Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 31
1.
Environ Int ; 170: 107603, 2022 12.
Article En | MEDLINE | ID: mdl-36335898

BACKGROUND: Structural analogues used to replace bisphenol A (BPA) since the introduction of new regulatory restrictions are considered emerging environmental toxicants and remain understudied with respect to their biological actions and health effects. Studies reveal a link between BPA exposure and vascular disease in human populations, whereas the vascular effects of BPA substitutes remain largely unknown. OBJECTIVES: To determine the effect of BPS, a commonly used BPA substitute, on redox balance, nitric oxide (NO) availability and microvascular NO-dependent dilation. METHODS: In human umbilical vein endothelial cells (HUVEC), production of reactive oxygen species (ROS) and NO after exposure to BPS was measured using fluorescent probes for DCFDA and DAF-FM diacetate, respectively. The contribution of endothelial NO synthase (eNOS) uncoupling to ROS generation was determined by measuring ROS in the presence or absence of an eNOS inhibitor (L-NAME) or eNOS co-factor, BH4, while the contribution of mitochondria-derived ROS was determined by treating cells with mitochondria-specific antioxidants prior to BPS exposure. Bioenergetic profiles were assessed using Seahorse extracellular flux analysis and mitochondria membrane polarization was measured with TMRE and JC-1 assays. In a mouse model of low dose BPS exposure, NO-mediated endothelial function was assessed in pressurized microvessels by inducing endothelium-dependent dilation in the presence or absence of L-NAME. RESULTS: BPS exposure (≥25 nM) reduced NO and increased ROS production in HUVEC, the latter corrected by treating cells with L-NAME or BH4. BPS exposure led to a loss of mitochondria membrane potential but had no impact on bioenergetic parameters except for a decrease in the spare respiratory capacity. Treatment of HUVEC with mitochondria-specific antioxidants abolished the effect of BPS on NO and ROS. NO-mediated vasodilation was impaired in male mice exposed to BPS. DISCUSSION: Exposure to BPS may promote cardiovascular disease by perturbing NO-mediated vascular homeostasis through the induction of oxidative stress.


Vascular Diseases , Male , Humans , Mice , Animals , Human Umbilical Vein Endothelial Cells , Vascular Diseases/chemically induced , Oxidative Stress
2.
Front Toxicol ; 4: 933572, 2022.
Article En | MEDLINE | ID: mdl-36310694

Background: Bisphenol S (BPS) is among the most commonly used substitutes for Bisphenol A (BPA), an endocrine disrupting chemical used as a plasticizer in the manufacture of polycarbonate plastics and epoxy resins. Bisphenols interfere with estrogen receptor (ER) signaling, which modulates vascular function through stimulation of nitric oxide (NO) production via endothelial nitric oxide synthase (eNOS). BPS can cross into the placenta and accumulates in the fetal compartment to a greater extent than BPA, potentially interfering with key developmental events. Little is known regarding the developmental impact of exposure to BPA substitutes, particularly with respect to the vasculature. Objective: To determine if prenatal BPS exposure influences vascular health in adulthood. Methods: At the time of mating, female C57BL/6 dams were administered BPS (250 nM) or vehicle control in the drinking water, and exposure continued during lactation. At 12-week of age, mesenteric arteries were excised from male and female offspring and assessed for responses to an endothelium-dependent (acetylcholine, ACh) and endothelium-independent (sodium nitroprusside, SNP) vasodilator. Endothelium-dependent dilation was measured in the presence or absence of L-NAME, an eNOS inhibitor. To further explore the role of NO and ER signaling, wire myography was used to assess ACh responses in aortic rings after acute exposure to BPS in the presence or absence of L-NAME or an ER antagonist. Results: Increased ACh dilation and increased sensitivity to Phe were observed in microvessels from BPS-exposed females, while no changes were observed in male offspring. Differences in ACh-induced dilation between control or BPS-exposed females were eliminated with L-NAME. Increased dilatory responses to ACh after acute BPS exposure were observed in aortic rings from female mice only, and differences were eliminated with inhibition of eNOS or inhibition of ER. Conclusion: Prenatal BPS exposure leads to persistent changes in endothelium-dependent vascular function in a sex-specific manner that appears to be modulated by interaction of BPS with ER signaling.

3.
Front Physiol ; 12: 752366, 2021.
Article En | MEDLINE | ID: mdl-35140625

Reactive oxygen species (ROS), such as superoxide anions and hydrogen peroxide, are reported to contribute to the dynamic regulation of contractility in various arterial preparations, however, the situation in pressurized, myogenically active resistance arteries is much less clear. In the present study, we have utilized established pharmacological inhibitors of NADPH oxidase activity to examine the potential contribution of ROS to intrinsic myogenic contractility in adult Sprague-Dawley rat resistance arteries and responses to vasoactive agents acting via the endothelium (i.e., acetylcholine, SKA-31) or smooth muscle (i.e., sodium nitroprusside, phenylephrine). In cannulated and pressurized cremaster skeletal muscle and middle cerebral arteries, the NOX inhibitors 2-acetylphenothiazine (2-APT) and VAS2870, selective for NOX1 and NOX2, respectively, evoked concentration-dependent inhibition of basal myogenic tone in a reversible and irreversible manner, respectively, whereas the non-selective inhibitor apocynin augmented myogenic contractility. The vasodilatory actions of 2-APT and VAS2870 occurred primarily via the vascular endothelium and smooth muscle, respectively. Functional responses to established endothelium-dependent and -independent vasoactive agents were largely unaltered in the presence of either 2-APT or apocynin. In cremaster arteries from Type 2 Diabetic (T2D) Goto-Kakizaki rats with endothelial dysfunction, treatment with either 2-APT or apocynin did not modify stimulus-evoked vasoactive responses, but did affect basal myogenic tone. These same NOX inhibitors produced robust inhibition of total NADPH oxidase activity in aortic tissue homogenates from control and T2D rats, and NOX isozymes 1, 2 and 4, along with superoxide dismutase 1, were detected by qPCR in cremaster arteries and aorta from both species. Based on the diverse effects that we observed for established, chemically distinct NOX inhibitors, the functional contribution of vascular NADPH oxidase activity to stimulus-evoked vasoactive signaling in myogenically active, small resistance arteries remains unclear.

4.
Naunyn Schmiedebergs Arch Pharmacol ; 391(5): 489-499, 2018 05.
Article En | MEDLINE | ID: mdl-29453527

Molecular investigations were performed in order to determine the major characteristics of voltage-gated Na+ channel ß-subunits in mouse vas deferens. The use of real-time quantitative PCR showed that the expression of Scn1b was significantly higher than that of other ß-subunit genes (Scn2b - Scn4b). Immunoreactivity of Scn1b proteins was also detected in the inner circular and outer longitudinal smooth muscle of mouse vas deferens. In whole-cell recordings, the actions of 4,9-anhydroTTX on voltage-gated Na+ current peak amplitude in myocytes (i.e., native INa) were compared with its inhibitory potency on recombinant NaV1.6 channels (expressed in HEK293 cells). A depolarizing rectangular voltage-pulse elicited a fast and transient inward native INa and recombinant NaV1.6 expressed in HEK293 cells (i.e., recombinant INa). The current decay of native INa was similar to the recombinant NaV1.6 current co-expressed with ß1-subunits. The current-voltage (I-V) relationships of native INa were similar to those of recombinant NaV1.6 currents co-expressed with ß1-subunits. Application of 4,9-anhydroTTX inhibited the peak amplitude of native INa (K i = 510 nM), recombinant INa (K i = 112 nM), and recombinant INa co-expressed with ß1-subunits (K i = 92 nM). The half-maximal (Vhalf) activation and inactivation of native INa values were similar to those observed in recombinant INa co-expressed with ß1-subunits. These results suggest that ß1-subunit proteins are likely to be expressed mainly in the smooth muscle layers of murine vas deferens and that 4,9-anhydroTTX inhibited not only native INa but also recombinant INa and recombinant INa co-expressed with ß1-subunits in a concentration-dependent manner.


Myocytes, Smooth Muscle/drug effects , Protein Subunits/physiology , Sodium Channel Blockers/pharmacology , Tetrodotoxin/analogs & derivatives , Vas Deferens/cytology , Voltage-Gated Sodium Channels/physiology , Animals , Cerebrum/drug effects , Cerebrum/physiology , HEK293 Cells , Humans , Male , Mice, Inbred BALB C , Myocytes, Smooth Muscle/physiology , Recombinant Proteins , Tetrodotoxin/pharmacology
5.
Microcirculation ; 24(3)2017 04.
Article En | MEDLINE | ID: mdl-28231612

OBJECTIVE: Mesenteric lymphatic vessel pumping, important to propel lymph and immune cells from the intestinal interstitium to the mesenteric lymph nodes, is compromised during intestinal inflammation. The objective of this study was to test the hypothesis that the pro-inflammatory cytokine TNF-α, is a significant contributor to the inflammation-induced lymphatic contractile dysfunction, and to determine its mode of action. METHODS: Contractile parameters were obtained from isolated rat mesenteric lymphatic vessels mounted on a pressure myograph after 24-hours incubation with or without TNF-α. Various inhibitors were administered, and quantitative real-time PCR, Western blotting, and immunofluorescence confocal imaging were applied to characterize the mechanisms involved in TNF-α actions. RESULTS: Vessel contraction frequency was significantly decreased after TNF-α treatment and could be restored by selective inhibition of NF-кB, iNOS, guanylate cyclase, and ATP-sensitive K+ channels. We further demonstrated that NF-кB inhibition also suppressed the significant increase in iNOS mRNA observed in TNF-α-treated lymphatic vessels and that TNF-α treatment favored the nuclear translocation of the p65 NF-κB subunit. CONCLUSIONS: These findings suggest that TNF-α decreases mesenteric lymphatic contractility by activating the NF-κB-iNOS signaling pathway. This mechanism could contribute to the alteration of lymphatic pumping reported in intestinal inflammation.


Lymphatic Vessels/physiopathology , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/pharmacology , Animals , Inflammation/metabolism , Mesentery/blood supply , Muscle Contraction/drug effects , Rats
6.
J Cereb Blood Flow Metab ; 37(1): 227-240, 2017 01.
Article En | MEDLINE | ID: mdl-26721393

The myogenic response of cerebral resistance arterial smooth muscle to intraluminal pressure elevation is a key physiological mechanism regulating blood flow to the brain. Rho-associated kinase plays a critical role in the myogenic response by activating Ca2+ sensitization mechanisms: (i) Rho-associated kinase inhibits myosin light chain phosphatase by phosphorylating its targeting subunit myosin phosphatase targeting subunit 1 (at T855), augmenting 20 kDa myosin regulatory light chain (LC20) phosphorylation and force generation; and (ii) Rho-associated kinase stimulates cytoskeletal actin polymerization, enhancing force transmission to the cell membrane. Here, we tested the hypothesis that abnormal Rho-associated kinase-mediated myosin light chain phosphatase regulation underlies the dysfunctional cerebral myogenic response of the Goto-Kakizaki rat model of type 2 diabetes. Basal levels of myogenic tone, LC20, and MYPT1-T855 phosphorylation were elevated and G-actin content was reduced in arteries of pre-diabetic 8-10 weeks Goto-Kakizaki rats with normal serum insulin and glucose levels. Pressure-dependent myogenic constriction, LC20, and myosin phosphatase targeting subunit 1 phosphorylation and actin polymerization were suppressed in both pre-diabetic Goto-Kakizaki and diabetic (18-20 weeks) Goto-Kakizaki rats, whereas RhoA, ROK2, and MYPT1 expression were unaffected. We conclude that abnormal Rho-associated kinase-mediated Ca2+ sensitization contributes to the dysfunctional cerebral myogenic response in the Goto-Kakizaki model of type 2 diabetes.


Actins/metabolism , Diabetes Mellitus, Type 2/physiopathology , Myosin-Light-Chain Phosphatase/antagonists & inhibitors , Vasoconstriction , Animals , Calcium/metabolism , Cerebral Arteries/physiopathology , Myosin-Light-Chain Phosphatase/metabolism , Phosphorylation , Polymerization , Rats , Rats, Inbred Strains , rho-Associated Kinases
7.
Biochem Pharmacol ; 97(3): 281-91, 2015 Oct 01.
Article En | MEDLINE | ID: mdl-26278977

The myogenic response of resistance arterioles and small arteries involving constriction in response to intraluminal pressure elevation and dilation on pressure reduction is fundamental to local blood flow regulation in the microcirculation. Integrins have garnered considerable attention in the context of initiating the myogenic response, but evidence indicative of mechanotransduction by integrin adhesions, for example established changes in tyrosine phosphorylation of key adhesion proteins, has not been obtained to substantiate this interpretation. Here, we evaluated the role of integrin adhesions and associated cellular signaling in the rat cerebral arterial myogenic response using function-blocking antibodies against α5ß1-integrins, pharmacological inhibitors of focal adhesion kinase (FAK) and Src family kinase (SFK), an ultra-high-sensitivity western blotting technique, site-specific phosphoprotein antibodies to quantify adhesion and contractile filament protein phosphorylation, and differential centrifugation to determine G-actin levels in rat cerebral arteries at varied intraluminal pressures. Pressure-dependent increases in the levels of phosphorylation of FAK (FAK-Y397, Y576/Y577), SFK (SFK-Y416; Y527 phosphorylation was reduced), vinculin-Y1065, paxillin-Y118 and phosphoinositide-specific phospholipase C-γ1 (PLCγ1)-Y783 were detected. Treatment with α5-integrin function-blocking antibodies, FAK inhibitor FI-14 or SFK inhibitor SU6656 suppressed the changes in adhesion protein phosphorylation, and prevented pressure-dependent phosphorylation of the myosin targeting subunit of myosin light chain phosphatase (MYPT1) at T855 and 20kDa myosin regulatory light chains (LC20) at S19, as well as actin polymerization that are necessary for myogenic constriction. We conclude that mechanotransduction by integrin adhesions and subsequent cellular signaling play a fundamental role in the cerebral arterial myogenic response.


Cerebral Arteries/metabolism , Integrin alpha5/metabolism , Muscle, Smooth, Vascular/metabolism , Signal Transduction , Vascular Resistance/physiology , Vasoconstriction/physiology , Animals , Arterial Pressure , Blotting, Western , In Vitro Techniques , Male , Myography , Phosphoproteins/metabolism , Phosphorylation , Pressure , Protein Kinases/metabolism , Rats, Sprague-Dawley
8.
J Biol Chem ; 289(30): 20939-52, 2014 Jul 25.
Article En | MEDLINE | ID: mdl-24914207

Our understanding of the molecular events contributing to myogenic control of diameter in cerebral resistance arteries in response to changes in intravascular pressure, a fundamental mechanism regulating blood flow to the brain, is incomplete. Myosin light chain kinase and phosphatase activities are known to be increased and decreased, respectively, to augment phosphorylation of the 20-kDa regulatory light chain subunits (LC20) of myosin II, which permits cross-bridge cycling and force development. Here, we assessed the contribution of dynamic reorganization of the actin cytoskeleton and thin filament regulation to the myogenic response and serotonin-evoked constriction of pressurized rat middle cerebral arteries. Arterial diameter and the levels of phosphorylated LC(20), calponin, caldesmon, cofilin, and HSP27, as well as G-actin content, were determined. A decline in G-actin content was observed following pressurization from 10 mm Hg to between 40 and 120 mm Hg and in three conditions in which myogenic or agonist-evoked constriction occurred in the absence of a detectable change in LC20 phosphorylation. No changes in thin filament protein phosphorylation were evident. Pressurization reduced G-actin content and elevated the levels of cofilin and HSP27 phosphorylation. Inhibitors of Rho-associated kinase and PKC prevented the decline in G-actin; reduced cofilin and HSP27 phosphoprotein content, respectively; and blocked the myogenic response. Furthermore, phosphorylation modulators of HSP27 and cofilin induced significant changes in arterial diameter and G-actin content of myogenically active arteries. Taken together, our findings suggest that dynamic reorganization of the cytoskeleton involving increased actin polymerization in response to Rho-associated kinase and PKC signaling contributes significantly to force generation in myogenic constriction of cerebral resistance arteries.


Actin Cytoskeleton/metabolism , Actin Depolymerizing Factors/metabolism , Cerebral Arterial Diseases/metabolism , G-Protein-Coupled Receptor Kinase 1/metabolism , HSP27 Heat-Shock Proteins/metabolism , Middle Cerebral Artery/metabolism , Protein Kinase C/metabolism , Actin Cytoskeleton/pathology , Animals , Calcium-Binding Proteins/metabolism , Cerebral Arterial Diseases/pathology , Constriction, Pathologic/metabolism , Constriction, Pathologic/pathology , Microfilament Proteins/metabolism , Middle Cerebral Artery/pathology , Phosphorylation , Rats , Rats, Sprague-Dawley , Calponins
9.
Biochem Biophys Res Commun ; 450(1): 440-6, 2014 Jul 18.
Article En | MEDLINE | ID: mdl-24928396

In studies of gene-ablated mice, activin signaling through activin type IIB receptors (ActRIIB) and Smad2 has been shown to regulate not only pancreatic ß cell mass but also insulin secretion. However, it still remains unclear whether gain of function of activin signaling is involved in the modulation of pancreatic ß cell mass and insulin secretion. To identify distinct roles of activin signaling in pancreatic ß cells, the Cre-loxP system was used to activate signaling through activin type IB receptor (ActRIB) in pancreatic ß cells. The resultant mice (pancreatic ß cell-specific ActRIB transgenic (Tg) mice; ActRIBCAßTg) exhibited a defect in glucose-stimulated insulin secretion (GSIS) and a progressive impairment of glucose tolerance. Patch-clamp techniques revealed that the activity of ATP-sensitive K(+) channels (KATP channels) was decreased in mutant ß cells. These results indicate that an appropriate level of activin signaling may be required for GSIS in pancreatic ß cells, and that activin signaling involves modulation of KATP channel activity.


Activin Receptors, Type I/metabolism , Activins/metabolism , Glucose/metabolism , Insulin Resistance/physiology , Insulin-Secreting Cells/metabolism , Insulin/metabolism , KATP Channels/physiology , Animals , Cells, Cultured , Insulin Secretion , Ion Channel Gating/physiology , Mice , Mice, Transgenic , Signal Transduction/physiology
10.
Br J Pharmacol ; 171(1): 145-57, 2014 Jan.
Article En | MEDLINE | ID: mdl-24117345

BACKGROUND AND PURPOSE: ATP-sensitive K(+)(K(ATP)) channels, which are composed of K(IR)6.x associated with sulphonylurea receptor (SUR) subunits, have been detected in native smooth muscle cells, but it is currently not known which of these is expressed in mouse vas deferens myocytes. EXPERIMENTAL APPROACH: Pharmacological and electrophysiological properties of K(ATP) channels in mouse vas deferens myocytes were investigated using patch clamp techniques. Molecular biological analyses were performed to examine the properties of these K(ATP) channels. KEY RESULTS: During conventional whole-cell recording, pinacidil elicited an inward current that was suppressed by glibenclamide, a sulfonylurea agent, and by U-37883A, a selective K(IR)6.1 blocker. When 0.3 mM ATP was added to the pipette solution, the peak amplitude of the pinacidil-induced current was much smaller than that recorded in its absence. When 3 mM UDP, GDP or ADP was included in the pipette solution, an inward current was elicited after establishment of the conventional whole-cell configuration, with potency order being UDP > GDP > ADP. These nucleoside diphosphate-induced inward currents were suppressed by glibenclamide. MCC-134, a SUR modulator, induced glibenclamide-sensitive K(ATP) currents that were similar to those induced by 100 µM pinacidil. In the cell-attached configuration, pinacidil activated channels with a conductance similar to that of K(IR)6.1. Reverse transcription PCR analysis revealed the expression of K(IR)6.1 and SUR2B transcripts and immunohistochemical studies indicated the presence of K(IR)6.1 and SUR2B proteins in the myocytes. CONCLUSIONS AND IMPLICATIONS: Our results indicate that native K(ATP) channels in mouse vas deferens myocytes are a heterocomplex of K(IR)6.1 channels and SUR2B subunits.


Adenosine Triphosphate/metabolism , KATP Channels/metabolism , Myocytes, Smooth Muscle/metabolism , Sulfonylurea Receptors/metabolism , Vas Deferens/metabolism , Adenosine Diphosphate/metabolism , Animals , Dose-Response Relationship, Drug , Guanosine Diphosphate/metabolism , HEK293 Cells , Humans , KATP Channels/antagonists & inhibitors , KATP Channels/genetics , Male , Membrane Potentials , Mice , Mice, Inbred BALB C , Myocytes, Smooth Muscle/drug effects , Potassium Channel Blockers/pharmacology , Sulfonylurea Receptors/antagonists & inhibitors , Sulfonylurea Receptors/genetics , Transfection , Uridine Diphosphate/metabolism , Vas Deferens/cytology , Vas Deferens/drug effects
11.
Diabetologia ; 57(1): 157-66, 2014 Jan.
Article En | MEDLINE | ID: mdl-24068386

AIMS/HYPOTHESIS: The TGF-ß superfamily of ligands provides important signals for the development of pancreas islets. However, it is not yet known whether the TGF-ß family signalling pathway is required for essential islet functions in the adult pancreas. METHODS: To identify distinct roles for the downstream components of the canonical TGF-ß signalling pathway, a Cre-loxP system was used to disrupt SMAD2, an intracellular transducer of TGF-ß signals, in pancreatic beta cells (i.e. Smad2ß knockout [KO] mice). The activity of ATP-sensitive K(+) channels (KATP channels) was recorded in mutant beta cells using patch-clamp techniques. RESULTS: The Smad2ßKO mice exhibited defective insulin secretion in response to glucose and overt diabetes. Interestingly, disruption of SMAD2 in beta cells was associated with a striking islet hyperplasia and increased pancreatic insulin content, together with defective glucose-responsive insulin secretion. The activity of KATP channels was decreased in mutant beta cells. CONCLUSIONS/INTERPRETATION: These results suggest that in the adult pancreas, TGF-ß signalling through SMAD2 is crucial for not only the determination of beta cell mass but also the maintenance of defining features of mature pancreatic beta cells, and that this involves modulation of KATP channel activity.


Hyperplasia/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Islets of Langerhans/metabolism , KATP Channels/metabolism , Smad2 Protein/metabolism , Animals , Electrophysiology , Female , Insulin Secretion , KATP Channels/genetics , Male , Mice , Mice, Knockout , Smad2 Protein/genetics
12.
Pflugers Arch ; 464(5): 493-502, 2012 Nov.
Article En | MEDLINE | ID: mdl-22986623

Patch-clamp experiments were performed to investigate the molecular properties of resurgent-like currents in single smooth muscle cells dispersed from mouse vas deferens, utilizing both Na(V)1.6-null mice (Na(V)1.6(-/-)), lacking the expression of the Scn8a Na(+) channel gene, and their wild-type littermates (Na(V)1.6(+/+)). Na(V)1.6 immunoreactivity was clearly visible in dispersed smooth muscle cells obtained from Na(V)1.6(+/+), but not Na(V)1.6(-/-), vas deferens. Following a depolarization to +30 mV from a holding potential of -70 mV (to produce maximal inactivation of the Na(+) current), repolarization to voltages between -60 and +20 mV elicited a tetrodotoxin (TTX)-sensitive inward current in Na(V)1.6(+/+), but not Na(V)1.6(-/-), vas deferens myocytes. The resurgent-like current in Na(V)1.6(+/+) vas deferens myocytes peaked at approximately -20 mV in the current-voltage relationship. The peak amplitude of the resurgent-like current remained at a constant level when the membrane potential was repolarized to -20 mV following the application of depolarizing rectangular pulses to more positive potentials than +20 mV. 4,9-Anhydrotetrodotoxin (4,9-anhydroTTX), a selective Na(V)1.6 blocking toxin, purified from a crude mixture of TTX analogues by LC-FLD techniques, reversibly suppressed the resurgent-like currents. ß-Pompilidotoxin, a voltage-gated Na(+) channel activator, evoked sustained resurgent-like currents in Na(V)1.6(+/+) but not Na(V)1.6(-/-) murine vas deferens myocytes. These results strongly indicate that, primarily, resurgent-like currents are generated as a result of Na(V)1.6 channel activity.


Action Potentials , Myocytes, Smooth Muscle/physiology , NAV1.6 Voltage-Gated Sodium Channel/physiology , Action Potentials/genetics , Animals , Insect Proteins/pharmacology , Male , Mice , Mice, Mutant Strains , NAV1.6 Voltage-Gated Sodium Channel/drug effects , NAV1.6 Voltage-Gated Sodium Channel/genetics , Tetrodotoxin/pharmacology , Vas Deferens/cytology , Voltage-Gated Sodium Channel Agonists/pharmacology , Voltage-Gated Sodium Channel Blockers/pharmacology , Wasp Venoms/pharmacology
13.
Hypertension ; 59(4): 877-84, 2012 Apr.
Article En | MEDLINE | ID: mdl-22353613

KCNQ4-encoded voltage-dependent potassium (Kv7.4) channels are important regulators of vascular tone that are severely compromised in models of hypertension. However, there is no information as to the role of these channels in responses to endogenous vasodilators. We used a molecular knockdown strategy, as well as pharmacological tools, to examine the hypothesis that Kv7.4 channels contribute to ß-adrenoceptor-mediated vasodilation in the renal vasculature and underlie the vascular deficit in spontaneously hypertensive rats. Quantitative PCR and immunohistochemistry confirmed gene and protein expression of KCNQ1, KCNQ3, KCNQ4, KCNQ5, and Kv7.1, Kv7.4, and Kv7.5 in rat renal artery. Isoproterenol produced concentration-dependent relaxation of precontracted renal arteries and increased Kv7 channel currents in isolated smooth muscle cells. Application of the Kv7 blocker linopirdine attenuated isoproterenol-induced relaxation and current. Isoproterenol-induced relaxations were also reduced in arteries incubated with small interference RNAs targeted to KCNQ4 that produced a ≈60% decrease in Kv7.4 protein level. Relaxation to isoproterenol and the Kv7 activator S-1 were abolished in arteries from spontaneously hypertensive rats, which was associated with ≈60% decrease in Kv7.4 abundance. This study provides the first evidence that Kv7 channels contribute to ß-adrenoceptor-mediated vasodilation in the renal vasculature and that abrogation of Kv7.4 channels is strongly implicated in the impaired ß-adrenoceptor pathway in spontaneously hypertensive rats. These findings may provide a novel pathogenic link between arterial dysfunction and hypertension.


Hypertension/physiopathology , KCNQ Potassium Channels/deficiency , Receptors, Adrenergic, beta/physiology , Renal Artery/physiology , Vasodilation/physiology , Adrenergic beta-Agonists/pharmacology , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Knockdown Techniques , Isoproterenol/pharmacology , KCNQ Potassium Channels/drug effects , KCNQ Potassium Channels/genetics , Male , RNA, Small Interfering/pharmacology , Rats , Rats, Inbred SHR , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/physiology , Vasodilation/drug effects
14.
J Cell Physiol ; 223(1): 234-43, 2010 Apr.
Article En | MEDLINE | ID: mdl-20054822

Patch-clamp experiments were performed to investigate the behavior of voltage-activated inward currents in vas deferens myocytes from Na(V)1.6-null mice (Na(V)1.6(-/-)) lacking the expression of the Na(+) channel gene, Scn8a, and their wild-type littermates (Na(V)1.6(+/+)). Immunohistochemistry confirmed expression of Na(V)1.6 in the muscle of Na(V)1.6(+/+), but not Na(V)1.6(-/-), vas deferens. PCR analysis revealed that the only beta(1)-subunit gene expressed in Na(V)1.6(+/+) vas deferens was Scn1b. In Na(V)1.6(+/+) myocytes, the threshold for membrane currents evoked by 20 msec voltage ramps (-100 mV to 60 mV) was -38.5 +/- 4.6 mV and this was shifted to a more positive potential (-31.2 +/- 4.9 mV) by tetrodotoxin (TTX). In Na(V)1.6(-/-) myocytes, the threshold was -30.4 +/- 3.4 mV and there was no TTX-sensitive current. The Na(+) current (I(Na)) in Na(V)1.6(+/+) myocytes had a bell-shaped current-voltage relationship that peaked at approximately -10 mV. Increasing the duration of the voltage ramps beyond 20 msec reduced the peak amplitude of I(Na). I(Na) displayed both fast (tau approximately 10 msec) and slow (tau approximately 1 sec) recovery from inactivation, the magnitude of the slow component increasing with the duration of the conditioning pulse (5-40 msec). During repetitive activation (5-40 msec pulses), I(Na) declined at stimulation frequencies > 0.5 Hz and at 10 Hz

Muscle, Smooth/metabolism , Myocytes, Smooth Muscle/metabolism , Nerve Tissue Proteins/metabolism , Sodium Channels/metabolism , Sodium/metabolism , Vas Deferens/metabolism , Animals , Cell Separation , Electric Stimulation , Immunohistochemistry , Ion Channel Gating , Kinetics , Male , Membrane Potentials , Mice , Mice, Inbred C3H , Mice, Knockout , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , Myocytes, Smooth Muscle/drug effects , NAV1.6 Voltage-Gated Sodium Channel , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Patch-Clamp Techniques , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sodium Channel Blockers/pharmacology , Sodium Channels/deficiency , Sodium Channels/genetics , Tetrodotoxin/pharmacology , Vas Deferens/cytology , Vas Deferens/drug effects , Voltage-Gated Sodium Channel beta-1 Subunit
15.
Br J Pharmacol ; 157(8): 1483-93, 2009 Aug.
Article En | MEDLINE | ID: mdl-19552689

BACKGROUND AND PURPOSE: The effects of veratridine, an alkaloid found in Liliaceae plants, on tetrodotoxin (TTX)-sensitive voltage-gated Na(+) channels were investigated in mouse vas deferens. EXPERIMENTAL APPROACH: Effects of veratridine on TTX-sensitive Na(+) currents (I(Na)) in vas deferens myocytes dispersed from BALB/c mice, homozygous mice with a null allele of Na(V)1.6 (Na(V)1.6(-/-)) and wild-type mice (Na(V)1.6(+/+)) were studied using patch-clamp techniques. Tension measurements were also performed to compare the effects of veratridine on phasic contractions in intact tissues. KEY RESULTS: In whole-cell configuration, veratridine had a concentration-dependent dual action on the peak amplitude of I(Na): I(Na) was enhanced by veratridine (1-10 microM), while higher concentrations (> or =30 microM) inhibited I(Na). Additionally, two membrane current components were evoked by veratridine, namely a sustained inward current during the duration of the depolarizing rectangular pulse and a tail current at the repolarization. Although veratridine caused little shift of the voltage dependence of the steady-state inactivation curve and the activation curve for I(Na), veratridine enhanced a non-inactivating component of I(Na). Veratridine caused no detectable contractions in vas deferens from Na(V)1.6(-/-) mice, although in tissues from Na(V)1.6(+/+) mice, veratridine (> or =3 microM) induced TTX-sensitive contractions. Similarly, no detectable inward currents were evoked by veratridine in Na(V)1.6(-/-) vas deferens myocytes, while veratridine elicited both the sustained and tail currents in cells taken from Na(V)1.6(+/+) mice. CONCLUSIONS AND IMPLICATIONS: These results suggest that veratridine possesses a dual action on I(Na) and that the veratridine-induced activation of contraction is induced by the activation of Na(V)1.6 channels.


Myocytes, Smooth Muscle/drug effects , Nerve Tissue Proteins/agonists , Sodium Channel Agonists , Vas Deferens/drug effects , Veratridine/pharmacology , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Ion Channel Gating , Male , Membrane Potentials/drug effects , Mice , Mice, Inbred BALB C , Muscle Contraction , Myocytes, Smooth Muscle/physiology , NAV1.6 Voltage-Gated Sodium Channel , Nerve Tissue Proteins/physiology , Patch-Clamp Techniques , Sodium Channels/physiology , Tetrodotoxin/pharmacology , Vas Deferens/physiology
16.
Naunyn Schmiedebergs Arch Pharmacol ; 379(5): 453-60, 2009 May.
Article En | MEDLINE | ID: mdl-19127357

Kurtoxin is described as a selective inhibitor of Ca(V)3.1. Using patch-clamp techniques, the modulatory effects of kurtoxin on tetrodotoxin-sensitive voltage-gated Na(+) currents (I(Na)) recorded from mouse vas deferens myocytes were investigated. Kurtoxin increased the peak amplitude of I(Na) between -40 and -30 mV, whilst inhibited the peak amplitude at more positive potentials than -10 mV, thereby demonstrating a dual action on the peak amplitude of I(Na). The time to reach the peak amplitude of I(Na) became significantly longer in the presence of kurtoxin in comparison with that of the controls. Kurtoxin also slowed the deactivation of I(Na) at more positive membrane potentials than -30 mV. Kurtoxin enhanced the total amount of electrical charge of I(Na) in a concentration-dependent manner. These results suggest that kurtoxin is a modulator of Na(V)1.6 in native freshly dispersed smooth muscle cells from mouse vas deferens as well as its action on Ca(V)3.1.


Membrane Transport Modulators/pharmacology , Myocytes, Smooth Muscle/drug effects , Nerve Tissue Proteins/antagonists & inhibitors , Scorpion Venoms/pharmacology , Vas Deferens/drug effects , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Ion Channel Gating/drug effects , Male , Membrane Potentials/drug effects , Mice , Mice, Inbred BALB C , Myocytes, Smooth Muscle/metabolism , NAV1.6 Voltage-Gated Sodium Channel , Nerve Tissue Proteins/biosynthesis , Patch-Clamp Techniques , Sodium Channels/biosynthesis , Tetrodotoxin/pharmacology , Vas Deferens/cytology , Vas Deferens/metabolism
17.
Eur J Pharmacol ; 605(1-3): 138-44, 2009 Mar 01.
Article En | MEDLINE | ID: mdl-19168048

Ca(2+) channel antagonists are useful to reduce the abnormal motility in patients with irritable bowel syndrome. We have therefore examined the effects of a newly synthesized antagonist AJG049, on voltage-dependent L-type Ca(2+) channels in gastric antrum. Intracellular recordings were made from sheets of the circular muscle layer of guinea-pig gastric antrum, with simultaneous measurement of spontaneous contraction activity, and the effects of AJG049 were studied. The effects of AJG049 on voltage-dependent Ba(2+) currents (I(Ba)) and the basal membrane currents at -70 mV in dispersed smooth muscle cells were also investigated by the use of conventional whole-cell patch-clamp techniques. Although AJG049 (100 nM) enhanced the peak amplitude of spontaneous contractions, high concentrations (>or=10 microM) had inhibitory effects. In whole-cell configuration, AJG049 (10 nM) reversibly enhanced the peak amplitude of I(Ba) in a voltage-dependent manner whilst high concentrations (>or=100 nM) suppressed the peak amplitude in a concentration- and voltage-dependent manner. AJG049 (300 nM) caused little shift in the activation curve at a holding potential of -70 mV although the voltage dependence of the steady-state inactivation was shifted to more negative potentials by 5 mV in its presence. AJG049 caused a delay of the recovery from the inactivated state of I(Ba). Furthermore, AJG049 reduced the amplitude of the basal membrane currents at -70 mV in a concentration-dependent manner. These results suggest that AJG049 possesses a dual action on voltage-dependent Ca(2+) channels in circular layer of guinea-pig antrum.


Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/drug effects , Oxazepines/pharmacology , Pyrrolidines/pharmacology , Animals , Calcium Channel Blockers/administration & dosage , Calcium Channels, L-Type/metabolism , Dose-Response Relationship, Drug , Female , Guinea Pigs , Male , Muscle Contraction/drug effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Oxazepines/administration & dosage , Patch-Clamp Techniques , Pyloric Antrum/drug effects , Pyloric Antrum/metabolism , Pyrrolidines/administration & dosage
18.
Am J Physiol Renal Physiol ; 296(1): F107-17, 2009 Jan.
Article En | MEDLINE | ID: mdl-18945825

The inwardly rectifying properties and molecular basis of ATP-sensitive K(+) channels (K(ATP) channels) have now been established for several cell types. However, these aspects of nonvascular smooth muscle K(ATP) channels still remain to be defined. In this study, we investigated the molecular basis of the pore of K(ATP) channels of pig urethral smooth muscle cells through a comparative study of the inwardly rectifying properties, conductance, and regulation by PKC of native and homo- and heteroconcatemeric recombinant Kir6.x channels coexpressed with sulfonylurea receptor subunit SUR2B in human embryonic kidney (HEK) 293 cells by the patch-clamp technique (conventional whole-cell and cell-attached modes). In conventional whole-cell clamp recordings, levcromakalim (> or = 1 microM) caused a concentration-dependent increase in current that demonstrated strong inward rectification at positive membrane potentials. In cell-attached mode, the unitary amplitude of levcromakalim-induced native and recombinant heteroconcatemeric Kir6.1-Kir6.2 K(ATP) channels also showed strong inward rectification at positive membrane potentials. Phorbol 12,13-dibutyrate, but not the inactive phorbol ester, 4alpha-phorbol 12,13-didecanoate, enhanced the activity of native and heteroconcatemeric K(ATP) channels at -50 mV. The conductance of the native channels at approximately 43 pS was consistent with that of heteroconcatemeric channels with a pore-forming subunit composition of (Kir6.1)(3)-(Kir6.2). RT-PCR analysis revealed the expression of Kir6.1 and Kir6.2 transcripts in pig urethral myocytes. Our findings provide the first evidence that the predominant K(ATP) channel expressed in pig urethral smooth muscle possesses a unique, heteromeric pore structure that differs from the homomeric Kir6.1 channels of vascular myocytes and is responsible for the differences in inward rectification, conductance, and PKC regulation exhibited by the channels in these smooth muscle cell types.


Myocytes, Smooth Muscle/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Protein Multimerization , Urethra/metabolism , Animals , Cromakalim/pharmacology , Dose-Response Relationship, Drug , Female , KATP Channels , Membrane Potentials/drug effects , Membrane Potentials/physiology , Myocytes, Smooth Muscle/cytology , Patch-Clamp Techniques , Phorbol 12,13-Dibutyrate/pharmacology , Potassium Channels, Inwardly Rectifying/drug effects , Protein Kinase C/metabolism , Swine , Urethra/cytology , Vasodilator Agents/pharmacology
19.
Naunyn Schmiedebergs Arch Pharmacol ; 378(6): 609-15, 2008 Dec.
Article En | MEDLINE | ID: mdl-18648774

(+/-)-Bay K 8644, a conventional racemic mixture of Bay K 8644, is widely used as an L-type Ca(2+) channel agonist. Although interactions between Bay K 8644 and cyclic nucleotide have been described, they have not been properly characterized. We have investigated whether two optical isomers of Bay K 8644 (i.e., R(+)- and S(-)-Bay K 8644) modify cyclic nucleotide (cAMP and cGMP)-induced inhibitory effects on nifedipine-sensitive voltage-dependent Ba(2+) currents (I (Ba)) recorded from guinea pig gastric myocytes. Conventional whole-cell recordings were used to compare the effects of R(+)-Bay K 8644 and S(-)-Bay K 8644 on I (Ba). S(-)-Bay K 8644 enhanced the peak amplitude of I (Ba) evoked by depolarizing pulses to +10 mV from a holding potential of -70 mV in a concentration-dependent manner (EC(50) = 32 nM), while R(+)-Bay K 8644 inhibited I (Ba) (IC(50) = 975 nM). When R(+)-Bay K 8644 (0.5 microM) was applied, I (Ba) was suppressed to 71 +/- 10% of control. In the presence of R(+)-Bay K 8644 (0.5 microM), additional application of forskolin and sodium nitroprusside (SNP) further inhibited I (Ba). Conversely, in the presence of S(-)-Bay K 8644 (0.5 microM), subsequent application of forskolin and SNP did not affect I (Ba). Similarly, in the presence of 0.5 microM S(-)-Bay K 8644, db-cAMP and 8-Br-cGMP had no effect on I (Ba). These results indicate that S(-)-Bay K 8644, but not R(+)-Bay K 8644, can prevent the inhibitory actions of two distinct cyclic nucleotide pathways on I (Ba) in gastric myocytes of the guinea pig antrum.


3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Barium/physiology , Bucladesine/pharmacology , Calcium Channel Agonists/pharmacology , Calcium Channels, L-Type/physiology , Cyclic GMP/analogs & derivatives , Pyloric Antrum/drug effects , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/chemistry , Animals , Bucladesine/metabolism , Calcium Channel Agonists/chemistry , Colforsin/metabolism , Colforsin/pharmacology , Cyclic GMP/metabolism , Cyclic GMP/pharmacology , Female , Guinea Pigs , In Vitro Techniques , Male , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Nitroprusside/metabolism , Nitroprusside/pharmacology , Patch-Clamp Techniques , Pyloric Antrum/metabolism , Stereoisomerism , Vasodilator Agents/metabolism , Vasodilator Agents/pharmacology
20.
Biophys J ; 94(8): 3340-51, 2008 Apr 15.
Article En | MEDLINE | ID: mdl-18192366

The biological and molecular properties of tetrodotoxin (TTX)-sensitive voltage-gated Na(+) currents (I(Na)) in murine vas deferens myocytes were investigated using patch-clamp techniques and molecular biological analyses. In whole-cell configuration, a fast, transient inward current was evoked in the presence of Cd(2+), and was abolished by TTX (K(d) = 11.2 nM), mibefradil (K(d) = 3.3 microM), and external replacement of Na(+) with monovalent cations (TEA(+), Tris(+), and NMDG(+)). The fast transient inward current was enhanced by veratridine, an activator of voltage-gated Na(+) channels, suggesting that the fast transient inward current was a TTX-sensitive I(Na). The values for half-maximal (V(half)) inactivation and activation of I(Na) were -46.3 mV and -26.0 mV, respectively. RT-PCR analysis revealed the expression of Scn1a, 2a, and 8a transcripts. The Scn8a transcript and the alpha-subunit protein of Na(V)1.6 were detected in smooth muscle layers. Using Na(V)1.6-null mice (Na(V)1.6(-/-)) lacking the expression of the Na(+) channel gene, Scn8a, I(Na) were not detected in dispersed smooth muscle cells from the vas deferens, while TTX-sensitive I(Na) were recorded in their wild-type (Na(V)1.6(+/+)) littermates. This study demonstrates that the molecular identity of the voltage-gated Na(+) channels responsible for the TTX-sensitive I(Na) in murine vas deferens myocytes is primarily Na(V)1.6.


Action Potentials/physiology , Ion Channel Gating/physiology , Membrane Potentials/physiology , Myocytes, Smooth Muscle/physiology , Sodium Channels/physiology , Sodium/metabolism , Vas Deferens/physiology , Animals , Cells, Cultured , Male , Mice , Mice, Inbred BALB C
...