Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Environ Sci Pollut Res Int ; 30(57): 120930-120944, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37945964

ABSTRACT

Substrates like sand or gravels and aquatic nutrient concentrations of rivers are highly heterogeneous, influencing the abundance of functional genes in epilithic biofilms where nitrification-denitrification processes take place. To analyze how the relative abundance of nitrifying/denitrifying genes and the associated microbes changes with the physical properties of substrates and aquatic concentrations of nutrients, this paper utilized metagenomics to comprehensively characterize these functional genes (i.e., amoA, hao, and nxrB involved in nitrification, and napA, narG, nirS, norB, and nosZ associated with denitrification) from epilithic biofilms collected along the Shitingjiang River in Southwest China and further obtained the relative abundance of major nitrifiers and denitrifiers. The results show that substrate size most significantly affects the relative abundance of hao and norB by altering the hydrodynamic conditions. In sampling sites with high heterogeneity in substrate size distribution, the relative abundance of most denitrifying genes is also higher. The carbon-nitrogen ratio negatively correlates with the relative abundance of all the nitrifying genes, while ammonium, total inorganic carbon, and total organic carbon concentrations positively affect the relative abundance of amoA and nxrB. As to the relative abundance of nitrifiers and denitrifiers, mainly belonging to phyla Proteobacteria and Actinobacteria, substrate heterogeneity and the aquatic concentrations of nutrients have greater influences than substrate size. Also, the substrate heterogeneity exerted positive influence on functional species of Pseudogemmobacter bohemicus and Paracoccus zhejiangensis. Considering the genes' functions and the dominant species linked to denitrification, nitrous oxide is more likely to occur in rivers with higher heterogeneity and larger substrates.


Subject(s)
Denitrification , Nitrification , Bacteria/genetics , Biofilms , Carbon , Nitrogen , Soil Microbiology
2.
Geohealth ; 6(8): e2022GH000607, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35991941

ABSTRACT

Stability of nitrogen cycle is a key indicator to aquatic health. In recent years, water and sediment inflows to the Three Gorges Reservoir (TGR) have changed significantly. To reveal the effects of such dramatic hydrological changes on aquatic nitrogen cycle, this paper at first analyzed the changing trends of water and suspended sediment discharges of TGR based on dynamic harmonic regression, and found that the intra-year distribution of water flow was significantly homogenized between flood and dry seasons, with the seasonal variations narrowed by 43.5%-69.9% during 2007-2016, while sediment concentration sharply dropped (the non-periodic term decreased by 1.48%-2.07%/month). Modified with the effects of sediment concentration variations on nitrification/denitrification rates, the proposed numerical model surprisingly showed that ammonia nitrogen and total nitrogen concentrations in TGR were insensitive to either water flow homogenization or sediment reduction, implying relative stability of microbial community related to nitrogen cycle, which is a positive sign for aquatic health. However, N2 emission varied more violently. The variation range of nitrogen gas (N2) emitted from TGR enlarged by 30% with the homogenization of water inflow from 2010 to 2016, while the annual total N2 emission decreased by 7% due to the reduction of sediment concentration, indicating quick response and strong adaption of the microbial N2 producing process to the environmental changes of TGR, which is beneficial for maintaining ecological functions related to nitrogen cycling. This work helps understanding nitrogen cycle of reservoirs experiencing dramatic changes in water and sediment inflows.

3.
Front Pharmacol ; 12: 658236, 2021.
Article in English | MEDLINE | ID: mdl-33935780

ABSTRACT

Purpose: The factor associated with IgA nephropathy (IgAN) is an abnormality of IgA known as galactose-deficient IgA1 (Gd-IgA1). The purpose of this study was to determine the molecular role played by miRNAs in the formation of Gd-IgA1 in IgAN and investigate the regulatory role of Astragaloside IV (AS-IV) in miRNAs. Patients and methods: Bioinformatics analysis, along with functional and mechanistic experiments, were used to investigate the relationship and function of miRNA, ß-1, 3-galactosyltransferase (C1GALT1), Gd-IgA1, and AS-IV. Analyses involved a series of tools, including quantitative real-time polymerase chain reaction (qRT-qPCR), Western blot, enzyme-linked immunosorbent assay (ELISA), Vicia Villosa lectin-binding assay (VVA), Cell counting kit-8 assay (CCK-8), and the dual-luciferase reporter assay. Results: miRNA screening and validation showed that miR-98-5p was significantly upregulated in the peripheral blood mononuclear cells (PBMCs) of pediatric patients with IgAN compared with patients diagnosed with mesangial proliferative glomerulonephritis (MsPGN) and immunoglobulin A vasculitis nephritis (IgAV-N), and healthy controls (p < 0.05). Experiments with the dual-luciferase reporter confirmed that miR-98-5p might target C1GALT1. The overexpression of miR-98-5p in DAKIKI cells decreased both the mRNA and protein levels of C1GALT1 and increased the levels of Gd-IgA1 levels; these effects were reversed by co-transfection with the C1GALT1 plasmid, and vice versa. In addition, AS-IV downregulated the levels of Gd-IgA1 level in DAKIKI cells by inhibiting miR-98-5p. Conclusions: Our results revealed that AS-IV could inhibit Gd-IgA1 secretion via miR-98-5p. Increased levels of miR-98-5p in pediatric IgAN patients might affect the glycosylation of IgA1 by targeting C1GALT1. In addition, our analyses suggest that the pathogenesis of IgAN may differ from that of IgAV-N. Collectively, these results provide significant insight into the pathogenesis of IgAN and identify a potential therapeutic target.

4.
Radiat Oncol ; 6: 144, 2011 Oct 26.
Article in English | MEDLINE | ID: mdl-22029423

ABSTRACT

BACKGROUND: Prostate cancer (PrCa) displays resistance to radiotherapy (RT) and requires radiotherapy dose escalation which is associated with greater toxicity. This highlights a need to develop radiation sensitizers to improve the efficacy of RT in PrCa. Ionizing radiation (IR) stimulates pathways of IR-resistance and survival mediated by the protein kinase Akt but it also activates the metabolic energy sensor and tumor suppressor AMP-Activated Protein Kinase (AMPK). Here, we examined the effects of the polyphenol resveratrol (RSV) on the IR-induced inhibition of cell survival, modulation of cell cycle and molecular responses in PrCa cells. METHODS: Androgen-insensitive (PC3), sensitive (22RV1) PrCa and PNT1A normal prostate epithelial cells were treated with RSV alone (2.5-10 µM) or in combination with IR (2-8 Gy). Clonogenic assays, cell cycle analysis, microscopy and immunoblotting were performed to assess survival, cell cycle progression and molecular responses. RESULTS: RSV (2.5-5 µM) inhibited clonogenic survival of PC3 and 22RV1 cells but not of normal prostate PNT1A cells. RSV specifically sensitized PrCa cells to IR, induced cell cycle arrest at G1-S phase and enhanced IR-induced nuclear aberrations and apoptosis. RSV enhanced IR-induced expression of DNA damage (γH2Ax) and apoptosis (cleaved-caspase 3) markers as well as of the cell cycle regulators p53, p21(cip1) and p27(kip1). RSV enhanced IR-activation of ATM and AMPK but inhibited basal and IR-induced phosphorylation of Akt. CONCLUSIONS: Our results suggest that RSV arrests cell cycle, promotes apoptosis and sensitizes PrCa cells to IR likely through a desirable dual action to activate the ATM-AMPK-p53-p21(cip1)/p27(kip1) and inhibit the Akt signalling pathways.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Prostatic Neoplasms/radiotherapy , Proto-Oncogene Proteins c-akt/metabolism , Stilbenes/pharmacology , TOR Serine-Threonine Kinases/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis , Cell Cycle , Cell Line, Tumor , Cell Survival , G1 Phase , Humans , Male , Microscopy, Fluorescence/methods , Radiation, Ionizing , Resveratrol
5.
J Thorac Oncol ; 6(3): 439-50, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21258249

ABSTRACT

INTRODUCTION: In this study, we investigated the effect of the 3-hydroxy-3-methylgutaryl-CoA reductase inhibitor lovastatin, as a sensitizer of lung cancer cells to ionizing radiation (IR). METHODS: A549 lung adenocarcinoma cells were treated with 0 to 50 µM lovastatin alone or in combination with 0 to 8 Gy IR and subjected to clonogenic survival and proliferation assays. To assess the mechanism of drug action, we examined the effects of lovastatin and IR on the epidermal growth factor (EGF) receptor and AMP-activated kinase (AMPK) pathways and on apoptotic markers and the cell cycle. RESULTS: Lovastatin inhibited basal clonogenic survival and proliferation of A549 cells and sensitized them to IR. This was reversed by mevalonate, the product of 3-hydroxy-3-methylgutaryl-CoA reductase. Lovastatin attenuated selectively EGF-induced phosphorylation of EGF receptor and Akt, and IR-induced Akt phosphorylation, in a mevalonate-sensitive fashion, without inhibition on extracellular signal-regulated kinase 1/2 phosphorylation by either stimulus. IR phosphorylated and activated the metabolic sensor and tumor suppressor AMPK, but lovastatin enhanced basal and IR-induced AMPK phosphorylation. The drug inhibited IR-induced expression of p53 and the cyclin-dependent kinase inhibitors p21(cip1) and p27(kip1), but caused a redistribution of cells from G1-S phase (control and radiated cells) and G2-M phase (radiated cells) of cell cycle into apoptosis. The latter was also evident by induction of nuclear fragmentation and cleavage of caspase 3 by lovastatin in both control and radiated cells. CONCLUSIONS: We suggest that lovastatin inhibits survival and induces radiosensitization of lung cancer cells through induction of apoptosis, which may be mediated by a simultaneous inhibition of the Akt and activation of the AMPK signaling pathways.


Subject(s)
Adenocarcinoma/radiotherapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lovastatin/pharmacology , Lung Neoplasms/radiotherapy , Radiation Tolerance/drug effects , Radiation, Ionizing , Adenocarcinoma/pathology , Apoptosis/drug effects , Apoptosis/radiation effects , Blotting, Western , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cytoprotection/drug effects , Cytoprotection/radiation effects , Fluorescent Antibody Technique , Humans , Lung Neoplasms/pathology , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
6.
Int J Radiat Oncol Biol Phys ; 78(1): 221-9, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20615625

ABSTRACT

PURPOSE: Adenosine monophosphate (AMP)-activated kinase (AMPK) is a molecular energy sensor regulated by the tumor suppressor LKB1. Starvation and growth factors activate AMPK through the DNA damage sensor ataxia-telangiectasia mutated (ATM). We explored the regulation of AMPK by ionizing radiation (IR) and its role as a target for radiosensitization of human cancer cells. METHODS AND MATERIALS: Lung, prostate, and breast cancer cells were treated with IR (2-8 Gy) after incubation with either ATM or AMPK inhibitors or the AMPK activator metformin. Then, cells were subjected to either lysis and immunoblotting, immunofluorescence microscopy, clonogenic survival assays, or cell cycle analysis. RESULTS: IR induced a robust phosphorylation and activation of AMPK in all tumor cells, independent of LKB1. IR activated AMPK first in the nucleus, and this extended later into cytoplasm. The ATM inhibitor KU-55933 blocked IR activation of AMPK. AMPK inhibition with Compound C or anti-AMPK alpha subunit small interfering RNA (siRNA) blocked IR induction of the cell cycle regulators p53 and p21(waf/cip) as well as the IR-induced G2/M arrest. Compound C caused resistance to IR, increasing the surviving fraction after 2 Gy, but the anti-diabetic drug metformin enhanced IR activation of AMPK and lowered the surviving fraction after 2 Gy further. CONCLUSIONS: We provide evidence that IR activates AMPK in human cancer cells in an LKB1-independent manner, leading to induction of p21(waf/cip) and regulation of the cell cycle and survival. AMPK appears to (1) participate in an ATM-AMPK-p21(waf/cip) pathway, (2) be involved in regulation of the IR-induced G2/M checkpoint, and (3) may be targeted by metformin to enhance IR responses.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Radiation Tolerance , AMP-Activated Protein Kinases/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins , Breast Neoplasms/metabolism , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Cyclin-Dependent Kinase Inhibitor p21/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA-Binding Proteins/metabolism , Enzyme Activation/drug effects , Enzyme Activation/radiation effects , Female , G2 Phase/drug effects , Humans , Lung Neoplasms/metabolism , Male , Metformin/pharmacology , Morpholines/pharmacology , Phosphorylation/radiation effects , Prostatic Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Pyrones/pharmacology , RNA, Small Interfering/pharmacology , Radiation Tolerance/drug effects , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism
7.
J Cardiovasc Pharmacol ; 50(4): 441-8, 2007 Oct.
Article in English | MEDLINE | ID: mdl-18049313

ABSTRACT

We compared the effects of genistein with its structural derivatives daidzein and equol on excitation of pulmonary artery and vein. The concentration of genistein necessary to inhibit contractions evoked by U46619 (1nM-100 microM) ranged from 10 to 100 microM. Genistein (55 microM) reduced KCl-responses by approximately 50% and essentially abolished those evoked by U46619. Daidzein was much less effective against either agonist, and equol was ineffective against U46619. A23187-evoked contractions were markedly reduced by all 3 isoflavones, but caffeine-evoked contractions were not. Using the Western blot technique, we found many proteins were tyrosine phosphorylated within 30 seconds after stimulation with U46619, reaching a peak at 120 seconds and then falling at 300 seconds. One band at 110 kD was increased nearly 300% above baseline, while 3 others ranging from 60 to 80 kD were more than doubled in intensity. Genistein had little effect on baseline levels of phosphorylation but largely prevented the U46619-induced change; daidzein was much less effective in this respect, and equol did not significantly affect this phosphorylation. We conclude that these isoflavones provide powerful tools in the study of excitation-contraction coupling of pulmonary vasculature and that inhibition of tyrosine kinase activity may be useful clinically against pulmonary hypertension.


Subject(s)
15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Phytoestrogens/pharmacology , Pulmonary Artery/drug effects , Pulmonary Veins/drug effects , Vasoconstriction/drug effects , Animals , Blotting, Western , Caffeine/pharmacology , Calcimycin/pharmacology , Cattle , Dose-Response Relationship, Drug , Equol , Genistein/pharmacology , In Vitro Techniques , Isoflavones/pharmacology , Muscle, Smooth, Vascular/drug effects , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Potassium Chloride/pharmacology , Protein-Tyrosine Kinases/metabolism , Proteins/chemistry , Proteins/metabolism , Pulmonary Artery/metabolism , Pulmonary Artery/physiology , Pulmonary Veins/metabolism , Pulmonary Veins/physiology , Tyrosine/metabolism , Vasoconstriction/physiology
8.
Article in English | MEDLINE | ID: mdl-17049219

ABSTRACT

We examined the responses to various isoprostane derivatives in bovine/human airway and pulmonary arteries. All biological activity of 15-F(2t)-IsoP was lost in its two major metabolites (15-keto-15-F(2t)-IsoP and 13,14-dihydro-15-keto-15-F(2t)-IsoP). We also examined the effects of several metabolites of 15-F(2t)-IsoP synthesized within our own laboratory-both epimers of 2,3-dinor-15-F(2t)-IsoP and of 2,3-dinor-5,6-dihydro-15-F(2t)-IsoP, as well as 20-carboxy-2,3,4,5-tetranor-15 oxo-5,6,13,14-tetrahydro-15-F(2t)-isoP)-finding none of these to have any substantial excitatory effect. Finally, several plant-derived isoprostanes ("phytoprostanes") synthesized within our laboratory elicited little or no excitatory response in these three pulmonary smooth muscle preparations. We conclude that, although isoprostane exhibit powerful constrictor effects on airway and pulmonary vascular smooth muscles, metabolic processing of those isoprostanes essentially abolishes those biological actions; also, the phytoprostanes lack any appreciable pharmacological activity on those smooth muscle preparations.


Subject(s)
Cyclopentanes/pharmacology , Fatty Acids, Unsaturated/pharmacology , Isometric Contraction/drug effects , Isoprostanes/pharmacology , Muscle, Smooth, Vascular/drug effects , Animals , Cattle , Dinoprost/analogs & derivatives , Dinoprost/metabolism , Dinoprost/pharmacology , Humans , In Vitro Techniques , Isoprostanes/metabolism , Pulmonary Artery/drug effects , Trachea/drug effects
9.
Am J Physiol Lung Cell Mol Physiol ; 291(6): L1177-84, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16815891

ABSTRACT

We previously reported the ability of isoprostanes to induce airway hyperresponsiveness (AHR). In this study, we examined the signaling mechanisms underlying that phenomenon with the standard muscle bath technique. Responses to a threshold concentration of carbachol (CCh, 3 x 10(-9) M) were significantly augmented by pretreatment for 20 min with 8-isoprostaglandin E(2) (15-E(2t)-IsoP, 10(-6) M): this AHR was obliterated in tissues pretreated with the selective Rho kinase (ROCK) inhibitor Y-27632 added 20 min before isoprostane, but not by cyclopiazonic acid (CPA). Increasing the CCh concentration to 3 x 10(-8) M (still considerably less than the half-maximally effective concentration of CCh) evoked larger contractions that were also augmented significantly by 15-E(2t)-IsoP: this AHR was completely abolished in tissues pretreated with CPA as well as those pretreated with Y-27632. We noted, however, that Y-27632 and CPA profoundly effect baseline tone and the cholinergic response per se, which confounds the interpretation of the data summarized above. We therefore modified the protocol by using combinations of CCh and blocker (CPA, Y-27632, or nifedipine) that were equieffective. In this way, we found that AHR could not be demonstrated under conditions in which Rho/ROCK signaling or Ca(2+) release was abolished (by Y-27632 and CPA, respectively). Likewise, other autacoids that act through G protein-coupled receptors via Rho/ROCK and Ca(2+) release (serotonin, histamine) mimicked this effect of isoprostane, whereas bradykinin did not. We conclude that isoprostane-induced AHR is mediated in part through an action on Rho/ROCK signaling. This novel finding may contribute to a better understanding of the mechanisms underlying AHR and asthma.


Subject(s)
Calcium/physiology , Intracellular Signaling Peptides and Proteins/physiology , Isoprostanes/pharmacology , Muscle, Smooth/physiology , Protein Serine-Threonine Kinases/physiology , Trachea/physiology , Amides/pharmacology , Animals , Bronchial Hyperreactivity/physiopathology , Carbachol/pharmacology , Cattle , Dinoprostone/pharmacology , Enzyme Inhibitors/pharmacology , In Vitro Techniques , Muscle, Smooth/drug effects , Pyridines/pharmacology , Respiratory System , Signal Transduction , rho-Associated Kinases
10.
Am J Physiol Lung Cell Mol Physiol ; 289(4): L574-82, 2005 Oct.
Article in English | MEDLINE | ID: mdl-15937065

ABSTRACT

Recently, we have shown that Rho and Rho-activated kinase (ROCK) may become activated by high-millimolar KCl, which had previously been widely assumed to act solely through opening of voltage-dependent Ca(2+) channels. In this study, we explored in more detail the relationship between membrane depolarization, Ca(2+) currents, and activation of Rho/ROCK in bovine tracheal smooth muscle. Ca(2+) currents began to activate at membrane voltages more positive than -40 mV and were maximally activated above 0 mV; at the same time, these underwent time- and voltage-dependent inactivation. Depolarizing intact tissues by KCl challenge evoked contractions that were blocked equally, and in a nonadditive fashion, by nifedipine or by the ROCK inhibitor Y-27632. Other agents that elevate intracellular calcium concentration ([Ca(2+)](i)) by pathways independent of G protein-coupled receptors, namely the SERCA-pump inhibitor cyclopiazonic acid and the Ca(2+) ionophore A-23187, evoked contractions that were also largely reduced by Y-27632. KCl directly increased Rho and ROCK activities in a concentration-dependent fashion that paralleled closely the effect of KCl on tone and [Ca(2+)](i), as well as the voltage-dependent Ca(2+) currents that were measured over the voltage ranges that are evoked by 0-120 mM KCl. Through the use of various pharmacological inhibitors, we ruled out roles for Ca(2+)/calmodulin-dependent CaM kinase II, protein kinase C, and protein kinase A in mediating the KCl-stimulated changes in tone and Rho/ROCK activities. In conclusion, Rho is activated by elevation of [Ca(2+)](i) (although the signal transduction pathway underlying this Ca(2+) dependence is still unclear) and possibly also by membrane depolarization per se.


Subject(s)
Calcium/metabolism , Muscle, Smooth/physiology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Trachea/physiology , rhoA GTP-Binding Protein/metabolism , Animals , Calcium Channels/drug effects , Calcium Channels/physiology , Cattle , Cells, Cultured , Intracellular Signaling Peptides and Proteins , Membrane Potentials/drug effects , Membrane Potentials/physiology , Muscle, Smooth/cytology , Potassium Chloride/pharmacology , Trachea/cytology , rho-Associated Kinases
11.
Am J Physiol Cell Physiol ; 288(3): C650-8, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15537706

ABSTRACT

Tonic contraction of corpus cavernosum smooth muscle cells (SMCs) maintains the flaccid state of the penis, and relaxation is initiated by nitric oxide (NO), leading to erection. Our aim was to investigate the effect of NO on the smooth muscle cellular response to adrenergic stimulation in corpus cavernosum. Fura-2 fluorescence was used to record intracellular Ca(2+) concentration ([Ca(2+)](i)) from freshly isolated SMCs from rat and human. Phenylephrine (PE) transiently elevated [Ca(2+)](i) in the presence and absence of extracellular Ca(2+), indicating release from intracellular stores. Whereas the NO donor S-nitroso-N-acetylpenicillamine (SNAP) with sildenafil citrate (SIL) caused no change in basal [Ca(2+)](i), the PE-induced rise of [Ca(2+)](i) was reversibly inhibited by 27 +/- 7% (n = 21, P < 0.005) in rat and by 55 +/- 15% (n = 9, P < 0.01) in human SMCs. SNAP and SIL also reduced the contractile response to PE. To investigate the mechanism, we applied mediators alone or in combination. The soluble guanylyl cyclase inhibitor ODQ reduced the effect of SNAP and SIL. SIL, cGMP analogs, and NO donors without SIL did not reduce the PE-induced rise of [Ca(2+)](i). However, the combination of 8-bromo-cGMP with SNAP reduced the Ca(2+) peak by 42 +/- 9% (n = 22, P < 0.01). Our results demonstrate that NO and cGMP act synergistically to reduce Ca(2+) release from intracellular stores. Reduction of intracellular Ca(2+) release may contribute to relaxation of the corpus cavernosum, leading to erection.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Cyclic GMP/metabolism , Muscle, Smooth/metabolism , Nitric Oxide/metabolism , Penis/anatomy & histology , 3',5'-Cyclic-GMP Phosphodiesterases/antagonists & inhibitors , Animals , Cells, Cultured , Fluorescent Dyes/metabolism , Fura-2/metabolism , Humans , Male , Muscle Contraction/physiology , Nitric Oxide Donors/metabolism , Piperazines/metabolism , Purines , Rats , Rats, Sprague-Dawley , Sildenafil Citrate , Sulfones , Vasodilator Agents/metabolism
12.
J Physiol ; 545(3): 829-36, 2002 12 15.
Article in English | MEDLINE | ID: mdl-12482889

ABSTRACT

Recent reports have suggested that different types of Ca(2+)-activated K(+) channels may be selectively expressed either in the vascular endothelial cells (ECs) or smooth muscle cells (SMCs) of a single artery. In this study, we directly compared mRNA, protein and functional expression of the high-conductance Ca(2+)-activated K(+) (BK(Ca)) channel between freshly isolated ECs and SMCs from bovine coronary arteries. Fresh ECs and SMCs were enzymatically isolated, and their separation verified by immunofluorescent detection of alpha-actin and platelet/endothelium cell adhesion molecule (PECAM) proteins, respectively. Subsequently, studies using a sequence-specific antibody directed against the pore-forming alpha-subunit of the BK(Ca) channel only detected its expression in the SMCs, whereas PECAM-positive ECs were devoid of the alpha-subunit protein. Additionally, multicell RT-PCR performed using cDNA derived from either SMCs or ECs only detected mRNA encoding the BK(Ca) alpha-subunit in the SMCs. Finally, whole-cell recordings of outward K(+) current detected a prominent iberiotoxin-sensitive BK(Ca) current in SMCs that was absent in ECs, and the BK(Ca) channel opener NS 1619 only enhanced K(+) current in the SMCs. Thus, bovine coronary SMCs densely express BK(Ca) channels whereas adjacent ECs in the same artery appear to lack the expression of the BK(Ca) channel gene. These findings indicate a cell-specific distribution of Ca(2+)-activated K(+) channels in SMCs and ECs from a single arterial site.


Subject(s)
Coronary Vessels/physiology , Endothelium, Vascular/physiology , Gene Expression , Potassium Channels, Calcium-Activated/genetics , Animals , Cattle , Coronary Vessels/cytology , Endothelium, Vascular/cytology , In Vitro Techniques , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits , Large-Conductance Calcium-Activated Potassium Channels , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/physiology , Patch-Clamp Techniques , Potassium Channels, Calcium-Activated/physiology , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...