Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Am J Physiol Cell Physiol ; 326(1): C1-C9, 2024 01 01.
Article in English | MEDLINE | ID: mdl-37955124

ABSTRACT

Airway smooth muscle (ASM) cells from mouse bronchus express a fast sodium current mediated by NaV1.7. We present evidence that this current is regulated by cAMP. ASM cells were isolated by enzymatic dispersal and studied using the whole cell patch clamp technique at room temperature. A fast sodium current, INa, was observed on holding cells under voltage clamp at -100 mV and stepping to -20 mV. This current was reduced in a concentration-dependent manner by denopamine (10 and 30 µM), a ß-adrenergic agonist. Forskolin (1 µM), an activator of adenylate cyclase, reduced the current by 35%, but 6-MB-cAMP (300 µM), an activator of protein kinase A (PKA), had no effect. In contrast, 8-pCPT-2-O-Me-cAMP-AM (007-AM, 10 µM), an activator of exchange protein directly activated by cAMP (Epac), reduced the current by 48%. The inhibitory effect of 007-AM was still observed in the presence of dantrolene (10 µM), an inhibitor of ryanodine receptors, and when cytosolic [Ca2+] was buffered by inclusion of 1,2-bis(o-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid, Sigma (BAPTA) (50 µM) in the pipette solution, suggesting that the inhibition of INa was not due to Ca2+-release from intracellular stores. When 007-AM was tested on the current-voltage relationship, it reduced the current at potentials from -30 to 0 mV, but had no effect on the steady-state activation curve. However, the steady-state inactivation V1/2, the voltage causing inactivation of 50% of the current, was shifted in the negative direction from -76.6 mV to -89.7 mV. These findings suggest that cAMP regulates INa in mouse ASM via Epac, but not PKA.NEW & NOTEWORTHY ß-adrenergic agonists are commonly used in inhalers to treat asthma and chronic obstructive pulmonary disease. These work by causing bronchodilation and reducing inflammation. The present study provides evidence that these drugs have an additional action, namely, to reduce sodium influx into airway smooth muscle cells via fast voltage-dependent channels. This may have the dual effect of promoting bronchodilation and reducing remodeling of the airways, which has a detrimental effect in these diseases.


Subject(s)
Cyclic AMP , Sodium , Mice , Animals , Sodium/metabolism , Cyclic AMP/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Myocytes, Smooth Muscle/metabolism , Adrenergic beta-Agonists
2.
Am J Physiol Cell Physiol ; 323(3): C749-C762, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35876287

ABSTRACT

Isolated smooth muscle cells (SMCs) from mouse bronchus were studied using the whole cell patch-clamp technique at ∼21°C. Stepping from -100 mV to -20 mV evoked inward currents of mean amplitude -275 pA. These inactivated (tau = 1.1 ms) and were abolished when external Na+ was substituted with N-Methyl-d-glucamine. In current-voltage protocols, current peaked at -10 mV and reversed between +20 and +30 mV. The V1/2s of activation and inactivation were -25 and -86 mV, respectively. The current was highly sensitive to tetrodotoxin (IC50 = 1.5 nM) and the NaV1.7 subtype-selective blocker, PF-05089771 (IC50 = 8.6 nM), consistent with NaV1.7 as the underlying pore-forming α subunit. Two NaV1.7-selective antibodies caused membrane-delineated staining of isolated SMC, as did a nonselective pan-NaV antibody. RT-PCR, performed on groups of ∼15 isolated SMCs, revealed transcripts for NaV1.7 in 7/8 samples. Veratridine (30 µM), a nonselective NaV channel activator, reduced peak current evoked by depolarization but induced a sustained current of 40 pA. Both effects were reversed by tetrodotoxin (100 nM). In tension experiments, veratridine (10 µM) induced contractions that were entirely blocked by atropine (1 µM). However, in the presence of atropine, veratridine was able to modulate the pattern of activity induced by a combination of U-46619 (a thromboxane A2 mimetic) and PGE2 (prostaglandin E2), by eliminating bursts in favor of sustained phasic contractions. These effects were readily reversed to control-like activity by tetrodotoxin (100 nM). In conclusion, mouse bronchial SMCs functionally express NaV1.7 channels that are capable of modulating contractile activity, at least under experimental conditions.


Subject(s)
Bronchi , Myocytes, Smooth Muscle , Animals , Atropine Derivatives/metabolism , Atropine Derivatives/pharmacology , Bronchi/metabolism , Mice , Myocytes, Smooth Muscle/metabolism , Sodium/metabolism , Tetrodotoxin/metabolism , Tetrodotoxin/pharmacology , Veratridine/metabolism , Veratridine/pharmacology
3.
Br J Pharmacol ; 179(5): 1082-1101, 2022 03.
Article in English | MEDLINE | ID: mdl-34767251

ABSTRACT

BACKGROUND AND PURPOSE: Corpus cavernosum smooth muscle (CCSM) exhibits phasic contractions that are coordinated by ion channels. Mouse models are commonly used to study erectile dysfunction, but there are few published electrophysiological studies of mouse CCSM. We describe the voltage-dependent sodium (NaV ) currents in mouse CCSM and investigate their function. EXPERIMENTAL APPROACH: We used electrophysiological, pharmacological and immunocytochemical methods to study the NaV currents in isolated CCSM cells from C57BL/6 mice. Tension measurements were carried out using crural sections of the corpus cavernosum in whole tissue. KEY RESULTS: Fast, voltage-dependent, sodium currents in mouse CCSM were induced by depolarising steps. Steady-state activation and inactivation curves revealed a window current between -60 and -30 mV. Two populations of NaV currents, 'TTX-sensitive' and 'TTX-insensitive', were identified. TTX-sensitive currents showed 48% block with the NaV channel subtype-specific blockers ICA-121431 (NaV 1.1-1.3), PF-05089771 (NaV 1.7) and 4,9-anhydro-TTX (NaV 1.6). TTX-insensitive currents were resistant to blockade by A803467, specific for NaV 1.8 channels. Immunocytochemistry confirmed expression of NaV 1.5 and NaV 1.4 in freshly dispersed CCSM cells. Veratridine, a NaV channel activator, reduced time-dependent inactivation of NaV currents and increased duration of evoked action potentials. Veratridine induced phasic contractions in CCSM strips, reversible with TTX and nifedipine but not KB-R7943. CONCLUSION AND IMPLICATIONS: There are fast, voltage-dependent, sodium currents in mouse CCSM. Stimulation of these currents increased contractility of CCSM in vitro, suggesting an involvement in detumescence and potentially providing a clinically relevant target in erectile dysfunction. Further work will be necessary to define its role.


Subject(s)
Erectile Dysfunction , Animals , Erectile Dysfunction/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle , Sodium/metabolism , Sodium Channel Blockers/metabolism , Sodium Channel Blockers/pharmacology , Tetrodotoxin/metabolism , Veratridine/metabolism
4.
Am J Physiol Cell Physiol ; 315(6): C818-C829, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30257105

ABSTRACT

Airway smooth muscle expresses abundant BKCa channels, but their role in regulating contractions remains controversial. This study examines the effects of two potent BKCa channel openers on agonist-induced phasic contractions in rabbit and mouse bronchi. First, we demonstrated the ability of 10 µM GoSlo-SR5-130 to activate BKCa channels in inside-out patches from rabbit bronchial myocytes, where it shifted the activation V1/2 by -88 ± 11 mV (100 nM Ca2+, n = 7). In mouse airway smooth muscle cells, GoSlo-SR5-130 dose dependently shifted V1/2 by 12-83 mV over a concentration range of 1-30 µM. Compound X, a racemic mixture of two enantiomers, reported to be potent BKCa channel openers, shifted V1/2 by 20-79 mV over a concentration range of 0.3-3 µM. In rabbit bronchial rings, exposure to histamine (1 µM) induced phasic contractions after a delay of ~35 min. These were abolished by GoSlo-SR5-130 (30 µM). Nifedipine (100 nM) and CaCCinhA01 (10 µM), a TMEM16A blocker, also abolished histamine-induced phasic contractions. In mouse bronchi, similar phasic contractions were evoked by exposure to U46619 (100 nM) and carbachol (100 nM). In each case, these were inhibited by concentrations of GoSlo-SR5-130 and compound X that shifted the activation V1/2 of BKCa channels in the order of -80 mV. In conclusion, membrane potential-dependent regulation of L-type Ca2+ channels appears to be important for histamine-, U46619-, and carbachol-induced phasic contractions in airway smooth muscle. Contractions can be abolished by BKCa channel openers, suggesting that these channels are potential targets for treating some causes of airway obstruction.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Membrane Potentials/drug effects , Myocytes, Smooth Muscle/metabolism , Animals , Anthraquinones/pharmacology , Bronchi/drug effects , Bronchi/growth & development , Bronchi/metabolism , Dose-Response Relationship, Drug , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/agonists , Membrane Potentials/genetics , Mice , Muscle Contraction/drug effects , Myocytes, Smooth Muscle/drug effects , Patch-Clamp Techniques , Rabbits , Sulfonic Acids/pharmacology
5.
BJU Int ; 121(6): 959-970, 2018 06.
Article in English | MEDLINE | ID: mdl-29211339

ABSTRACT

OBJECTIVE: To test if carbachol (CCh)-evoked Ca2+ oscillations in freshly isolated murine detrusor myocytes are affected by ß3-adrenoceptor (ß-AR) modulators. MATERIALS AND METHODS: Isometric tension recordings were made from strips of murine detrusor, and intracellular Ca2+ measurements were made from isolated detrusor myocytes using confocal microscopy. Transcriptional expression of ß-AR sub-types in detrusor strips and isolated detrusor myocytes was assessed using reverse transcriptase-polymerase chain reaction (RT-PCR) and real-time quantitative PCR (qPCR). Immunocytochemistry experiments, using a ß3-AR selective antibody, were performed to confirm that ß3-ARs were present on detrusor myocytes. RESULTS: The RT-PCR and qPCR experiments showed that ß1-, ß2- and ß3-AR were expressed in murine detrusor, but that ß3-ARs were the most abundant sub-type. The selective ß3-AR agonist BRL37344 reduced the amplitude of CCh-induced contractions of detrusor smooth muscle. These responses were unaffected by addition of the BK channel blocker iberiotoxin. BRL37344 also reduced the amplitude of CCh-induced Ca2+ oscillations in freshly isolated murine detrusor myocytes. This effect was mimicked by CL316,243, another ß3-AR agonist, and inhibited by the ß3-AR antagonist L748,337, but not by propranolol, an antagonist of ß1- and ß2-ARs. BRL37344 did not affect caffeine-evoked Ca2+ transients or L-type Ca2+ current in isolated detrusor myocytes. CONCLUSION: Inhibition of cholinergic-mediated contractions of the detrusor by ß3-AR agonists was associated with a reduction in Ca2+ oscillations in detrusor myocytes.


Subject(s)
Adrenergic beta-3 Receptor Agonists/pharmacology , Carbachol/pharmacology , Cholinergic Agonists/pharmacology , Muscle Cells/drug effects , Adrenergic beta-Agonists/pharmacology , Animals , Calcium/metabolism , Ethanolamines/pharmacology , Female , Male , Mice, Inbred C57BL , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Peptides/pharmacology , Urinary Bladder/drug effects
6.
Eur J Pharmacol ; 814: 216-225, 2017 Nov 05.
Article in English | MEDLINE | ID: mdl-28822854

ABSTRACT

Cyclic nucleotide-gated (CNG) channels are non-selective cation channels that mediate influx of extracellular Na+ and Ca2+ in various cell types. L-cis-Diltiazem, a CNG channel blocker, inhibits contraction of urethral smooth muscle (USM), however the mechanisms underlying this effect are still unclear. We investigated the possibility that CNG channels contribute to spontaneous pacemaker activity in freshly isolated interstitial cells of Cajal (ICC) isolated from the rabbit urethra (RUICC). Using immunocytochemistry, we found intense CNG1-immunoreactivity in vimentin-immunoreactive RUICC, mainly within patches of the cellular body and processes. In contrast, α-actin immunoreactive smooth muscle cells (SMC) did not show significant reactivity to a specific CNGA1 antibody. Freshly isolated RUICC, voltage clamped at -60mV, developed spontaneous transient inward currents (STICs) that were inhibited by L-cis-Diltiazem (50µM). Similarly, L-cis-Diltiazem (50µM) also inhibited Ca2+ waves in isolated RUICC, recorded using a Nipkow spinning disk confocal microscope. L-cis-Diltiazem (50µM) did not affect caffeine (10mM)-induced Ca2+ transients, but significantly reduced phenylephrine-evoked Ca2+ oscillations and inward currents in in RUICC. L-type Ca2+ current amplitude in isolated SMC was reduced by ~18% in the presence of L-cis-Diltiazem (50µM), however D-cis-Diltiazem, a recognised L-type Ca2+ channel blocker, abolished L-type Ca2+ current but did not affect Ca2+ waves or STICs in RUICC. These results indicate that the effects of L-cis-diltiazem on rabbit USM could be mediated by inhibition of CNG1 channels that are present in urethral ICC and therefore CNG channels contribute to spontaneous activity in these cells.


Subject(s)
Cyclic Nucleotide-Gated Cation Channels/metabolism , Interstitial Cells of Cajal/metabolism , Urethra/cytology , Animals , Calcium Signaling/drug effects , Diltiazem/pharmacology , Female , Interstitial Cells of Cajal/cytology , Interstitial Cells of Cajal/drug effects , Male , Rabbits
7.
Pflugers Arch ; 469(11): 1443-1455, 2017 11.
Article in English | MEDLINE | ID: mdl-28733893

ABSTRACT

Interstitial cells of Cajal (ICC) isolated from the rabbit urethra exhibit Ca2+-activated Cl- currents (I ClCa) that are important for the development of urethral tone. Here, we examined if TMEM16A (ANO1) contributed to this activity by examining the effect of "new-generation" TMEM16A inhibitors, CACCinh-A01 and T16Ainh-A01, on I ClCa recorded from freshly isolated rabbit urethral ICC (RUICC) and on contractions of intact strips of rabbit urethra smooth muscle. Real-time quantitative PCR experiments demonstrated that TMEM16A was highly expressed in rabbit urethra smooth muscle, in comparison to TMEM16B and TMEM16F. Single-cell RT-PCR experiments revealed that only TMEM16A was expressed in freshly isolated RUICC. Depolarization-evoked I ClCa in isolated RUICC, recorded using voltage clamp, were inhibited by CACCinh-A01 and T16Ainh-A01 with IC50 values of 1.2 and 3.4 µM, respectively. Similarly, spontaneous transient inward currents (STICs) recorded from RUICC voltage clamped at -60 mV and spontaneous transient depolarizations (STDs), recorded in current clamp, were also inhibited by CACCinh-A01 and T16Ainh-A01. In contrast, spontaneous Ca2+ waves in isolated RUICC were only partially reduced by CACCinh-A01 and T16Ainh-A01. Finally, neurogenic contractions of strips of rabbit urethra smooth muscle (RUSM), evoked by electric field stimulation (EFS), were also significantly reduced by CACCinh-A01 and T16Ainh-A01. These data are consistent with the idea that TMEM16A is involved with CACCs in RUICC and in contraction of rabbit urethral smooth muscle.


Subject(s)
Anoctamin-1/antagonists & inhibitors , Calcium/metabolism , Chlorides/metabolism , Interstitial Cells of Cajal/drug effects , Membrane Potentials/drug effects , Urethra/drug effects , Animals , Cells, Cultured , Chloride Channels/metabolism , Female , Interstitial Cells of Cajal/metabolism , Male , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Rabbits , Urethra/metabolism
8.
J Urol ; 196(6): 1796-1808, 2016 12.
Article in English | MEDLINE | ID: mdl-27287524

ABSTRACT

PURPOSE: Muscarinic receptor mediated contractions of the detrusor rely on Ca2+ influx through voltage-gated Ca2+ channels but to our knowledge the mechanism linking stimulation of M3Rs to the activation of voltage dependent Ca2+ channels has not been established. TRPC4 channels are receptor operated cation channels that couple muscarinic receptor activation to depolarization of intestinal smooth muscle cells, voltage-activated Ca2+ influx and contraction. We investigated whether TRPC4 channels are involved in cholinergic mediated contractions of the detrusor. MATERIALS AND METHODS: Isometric tension recordings were made on strips of murine detrusor and intracellular Ca2+ measurements were made on isolated detrusor myocytes using confocal microscopy. Transcriptional expression of TRPC and IP3R subtypes in intact detrusor strips and isolated detrusor myocytes was assessed using reverse transcriptase-polymerase chain reaction. RESULTS: Cholinergic stimulation of the detrusor induced by electrical field stimulation or exogenous application of carbachol or neostigmine evoked contractions consisting of a transient plus a tonic response, which was blocked by ML204, an inhibitor of TRPC4 channels. A phasic oscillatory component was blocked by the IP3R inhibitor 2-APB. Carbachol evoked reproducible Ca2+ responses in isolated detrusor myocytes, consisting of an initial Ca2+ transient followed by Ca2+ oscillations. ML204 inhibited the initial Ca2+ transient whereas 2-APB inhibited the Ca2+ oscillations. Reverse transcriptase-polymerase chain reaction experiments showed that TRPC4ß, TRPC6 and IP3R1 were selectively expressed in isolated detrusor myocytes. Control experiments demonstrated that ML204 did not affect L-type Ca2+ or BK current amplitude, caffeine induced Ca2+ transients or KCl induced contractions of the detrusor. CONCLUSIONS: Muscarinic receptor mediated contractions of the detrusor involve the activation of TRPC4ß channels.


Subject(s)
Muscle Contraction/physiology , Muscle, Smooth/physiology , Receptors, Muscarinic/physiology , TRPC Cation Channels/physiology , Urinary Bladder/physiology , Animals , Female , Male , Mice, Inbred C57BL
9.
Channels (Austin) ; 8(4): 308-20, 2014.
Article in English | MEDLINE | ID: mdl-24642630

ABSTRACT

Recent studies have shown that transmembrane protein 16 A (TMEM16A) is a subunit of calcium-activated chloride channels (CACCs). Pharmacological agents have been used to probe the functional role of CACCs, however their effect on TMEM16A currents has not been systematically investigated. In the present study, we characterized the voltage and concentration-dependent effects of 2 traditional CACC inhibitors (niflumic acid and anthracene-9-carboxcylic acid) and 2 novel CACC / TMEM16A inhibitors (CACC(inh)A01 and T16A(inh)A01) on TMEM16A currents. The whole cell patch clamp technique was used to record TMEM16A currents from HE K 293 cells that stably expressed human TMEM16A. Niflumic acid, A-9-C, CACC(inh)A01 and T16A(inh)A01 inhibited TMEM16A currents with IC50 values of 12, 58, 1.7 and 1.5 µM, respectively, however, A-9-C and niflumic acid were less efficacious at negative membrane potentials. A-9-C and niflumic acid reduced the rate of TMEM16A tail current deactivation at negative membrane potentials and A-9-C (1 mM) enhanced peak TMEM16A tail current amplitude. In contrast, the inhibitory effects of CACC(inh)A01 and T16A(inh)A01 were independent of voltage and they did not prolong the rate of TMEM16A tail current deactivation. The effects of niflumic acid and A-9-C on TMEM16A currents were similar to previous observations on CACCs in vascular smooth muscle, strengthening the hypothesis that they are encoded by TMEM16A. However, CACC(inh)A01 and T16A(inh)A01 were more potent inhibitors of TMEM16A channels and their effects were not diminished at negative membrane potentials making them attractive candidates to interrogate the functional role of TMEM16A channels in future studies.


Subject(s)
Chloride Channels/metabolism , Ion Channel Gating/drug effects , Neoplasm Proteins/metabolism , Niflumic Acid/pharmacology , Anoctamin-1 , Anthracenes/pharmacology , HEK293 Cells , Humans , Kinetics , Patch-Clamp Techniques , Pyrimidines/pharmacology , Thiazoles/pharmacology
10.
Am J Physiol Cell Physiol ; 305(6): C609-22, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23804200

ABSTRACT

We used the perforated patch-clamp technique at 37°C to investigate the mechanisms underlying the activation of a transient large-conductance K(+) (tBK) current in rabbit urethral smooth muscle cells. The tBK current required an elevation of intracellular Ca(2+), resulting from ryanodine receptor (RyR) activation via Ca(2+)-induced Ca(2+) release, triggered by Ca(2+) influx through L-type Ca(2+) (CaV) channels. Carbachol inhibited tBK current by reducing Ca(2+) influx and Ca(2+) release and altered the shape of spike complexes recorded under current-clamp conditions. The tBK currents were blocked by iberiotoxin and penitrem A (300 and 100 nM, respectively) and were also inhibited when external Ca(2+) was removed or the CaV channel inhibitors nifedipine (10 µM) and Cd(2+) (100 µM) were applied. The tBK current was inhibited by caffeine (10 mM), ryanodine (30 µM), and tetracaine (100 µM), suggesting that RyR-mediated Ca(2+) release contributed to the activation of the tBK current. When IP3 receptors (IP3Rs) were blocked with 2-aminoethoxydiphenyl borate (2-APB, 100 µM), the amplitude of the tBK current was not reduced. However, when Ca(2+) release via IP3Rs was evoked with phenylephrine (1 µM) or carbachol (1 µM), the tBK current was inhibited. The effect of carbachol was abolished when IP3Rs were blocked with 2-APB or by inhibition of muscarinic receptors with the M3 receptor antagonist 4-diphenylacetoxy-N-methylpiperidine methiodide (1 µM). Under current-clamp conditions, bursts of action potentials could be evoked with depolarizing current injection. Carbachol reduced the number and amplitude of spikes in each burst, and these effects were reduced in the presence of 2-APB. In the presence of ryanodine, the number and amplitude of spikes were also reduced, and carbachol was without further effect. These data suggest that IP3-generating agonists can modulate the electrical activity of rabbit urethral smooth muscle cells and may contribute to the effects of neurotransmitters on urethral tone.


Subject(s)
Inositol 1,4,5-Trisphosphate Receptors/metabolism , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Muscle, Smooth/metabolism , Myocytes, Smooth Muscle/metabolism , Urethra/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Animals , Boron Compounds/pharmacology , Caffeine/pharmacology , Calcium/metabolism , Calcium Channels, L-Type/metabolism , Carbachol/pharmacology , Female , Large-Conductance Calcium-Activated Potassium Channels/agonists , Large-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Male , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , Mycotoxins/pharmacology , Myocytes, Smooth Muscle/drug effects , Peptides/pharmacology , Potassium/metabolism , Rabbits , Receptor, Muscarinic M3/metabolism , Ryanodine/pharmacology , Ryanodine Receptor Calcium Release Channel/metabolism , Tetracaine/pharmacology , Urethra/drug effects
11.
J Urol ; 186(2): 745-52, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21683405

ABSTRACT

PURPOSE: Adenosine triphosphate is capable of relaxing and contracting urethral smooth muscle. The mechanisms responsible for the relaxing effects of adenosine triphosphate have been well studied but those involved in the contractile response are still unclear. We investigated the contributions of interstitial cells of Cajal and smooth muscle cells to nerve mediated, adenosine triphosphate dependent contractions of urethral smooth muscle. MATERIALS AND METHODS: Tension recordings were made from strips of rabbit urethral smooth muscle. Recordings were made of membrane potential and ionic currents from freshly isolated smooth muscle cells and interstitial cells of Cajal using the patch clamp technique. RESULTS: Stimulating intramural nerves in urethral smooth muscle yielded contractions that were inhibited by the broad spectrum P2 receptor inhibitor pyridoxal-phosphate-6-azophenyl-2',4'-disulfonate and the P2X receptor agonist α,ß-methylene adenosine triphosphate but not by the P2Y receptor antagonist MRS2500. When studied under voltage clamp at a holding potential of -60 mV, interstitial cells of Cajal showed spontaneous transient inward currents that were increased in frequency by adenosine triphosphate but not by α,ß-methylene adenosine triphosphate. In contrast, smooth muscle cells were quiescent but responded to adenosine triphosphate and α,ß-methylene adenosine triphosphate by producing a single transient inward current. Currents evoked by adenosine triphosphate in smooth muscle cells were inhibited by α,ß-methylene adenosine triphosphate, pyridoxal-phosphate-6-azophenyl-2',4'-disulfonate and suramin, and by a decrease in extracellular Na+ from 130 to 13 mM. CONCLUSIONS: Stimulating purinergic nerves in rabbit urethral smooth muscle induces contractions via the activation of P2X receptors on smooth muscle cells.


Subject(s)
Muscle Contraction , Muscle, Smooth/physiology , Receptors, Purinergic P2X/physiology , Urethra/physiology , Animals , Electrophysiological Phenomena , Female , In Vitro Techniques , Male , Muscle, Smooth/innervation , Rabbits , Urethra/innervation
12.
J Urol ; 183(2): 801-11, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20022059

ABSTRACT

PURPOSE: Adenosine triphosphate is thought to be an important neurotransmitter in urethral smooth muscle but its physiological role is still unclear. We characterized the effects of adenosine triphosphate on contractile and pacemaker activity in rabbit urethral smooth muscle. MATERIALS AND METHODS: Tension recordings were made from strips of rabbit proximal urethral smooth muscle. Membrane currents from freshly isolated smooth muscle cells and interstitial cells of Cajal were recorded using the patch clamp technique. Intracellular Ca(2+) was measured using confocal microscopy. RESULTS: Exogenous application of adenosine triphosphate (10 microM) evoked robust contractions that were inhibited by the type 2 purinergic receptor blocker suramin (100 microM) and the selective type 2 purinergic Y1 receptor antagonist MRS2500 (Tocris Bioscience, Ellisville, Missouri) (100 nM). Application of the type 2 purinergic Y receptor agonist 2-MeSADP (1 microM) mimicked the effects of adenosine triphosphate. When smooth muscle cells were studied under voltage clamp at -60 mV, adenosine triphosphate evoked a large single inward current (greater than 1.2 nA) but 2-MeSADP produced a small current (about 16 pA). In contrast, when interstitial cells of Cajal were held at -60 mV, they showed spontaneous transient inward currents that were increased in frequency by adenosine triphosphate and 2-MeSADP. These excitatory effects were inhibited by suramin and MRS2500. Interstitial cells of Cajal showed spontaneous Ca(2+) waves that were increased in frequency by adenosine triphosphate and 2-MeSADP. These effects were also inhibited by suramin and MRS2500. CONCLUSIONS: Contractile effects of adenosine triphosphate in urethral smooth muscle are mediated by the activation of type 2 purinergic Y receptors on interstitial cells of Cajal.


Subject(s)
Adenosine Triphosphate/physiology , Interstitial Cells of Cajal/physiology , Muscle Contraction/physiology , Muscle, Smooth/physiology , Urethra/physiology , Adenosine Triphosphate/pharmacology , Animals , Female , Interstitial Cells of Cajal/drug effects , Male , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Rabbits , Urethra/drug effects
13.
J Physiol ; 586(19): 4631-42, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18703577

ABSTRACT

Interstitial cells of Cajal (ICC) isolated from the rabbit urethra exhibit pacemaker activity that results from spontaneous Ca(2+) waves. The purpose of this study was to investigate if this activity was influenced by Ca(2+) uptake into mitochondria. Spontaneous Ca(2+) waves were recorded using a Nipkow spinning disk confocal microscope and spontaneous transient inward currents (STICs) were recorded using the whole-cell patch clamp technique. Disruption of the mitochondrial membrane potential with the electron transport chain inhibitors rotenone (10 microm) and antimycin A (5 microm) abolished Ca(2+) waves and increased basal Ca(2+) levels. Similar results were achieved when mitochondria membrane potential was collapsed using the protonophores FCCP (0.2 microm) and CCCP (1 microm). Spontaneous Ca(2+) waves were not inhibited by the ATP synthase inhibitor oligomycin (1 microm), suggesting that these effects were not attributable to an effect on ATP levels. STICs recorded under voltage clamp at -60 mV were also inhibited by CCCP and antimycin A. Dialysis of cells with the mitochondrial uniporter inhibitor RU360 (10 microm) also inhibited STICS. Stimulation of Ca(2+) uptake into mitochondria using the plant flavonoid kaempferol (10 microm) induced a series of propagating Ca(2+) waves. The kaempferol-induced activity was inhibited by application of caffeine (10 mm) or removal of extracellular Ca(2+), but was not significantly affected by the IP(3) receptor blocker 2-APB (100 microm). These data suggest that spontaneous Ca(2+) waves in urethral ICC are regulated by buffering of cytoplasmic Ca(2+) by mitochondria.


Subject(s)
Calcium/metabolism , Mitochondria/metabolism , Muscle, Smooth/metabolism , Urethra/metabolism , Animals , Antimycin A/pharmacology , Caffeine/pharmacology , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacology , Cells, Cultured , Central Nervous System Stimulants/pharmacology , Electron Transport Complex I/antagonists & inhibitors , Electron Transport Complex III/antagonists & inhibitors , Female , Kaempferols/pharmacology , Male , Membrane Potentials , Mitochondria/drug effects , Muscle, Smooth/cytology , Patch-Clamp Techniques , Rabbits , Rotenone/pharmacology , Urethra/cytology
14.
Am J Physiol Cell Physiol ; 289(3): C625-32, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16100389

ABSTRACT

The aim of the present study was to investigate the properties and role of capacitative Ca(2+) entry (CCE) in interstitial cells (IC) isolated from the rabbit urethra. Ca(2+) entry in IC was larger in cells with depleted intracellular Ca(2+) stores compared with controls, consistent with influx via a CCE pathway. The nonselective Ca(2+) entry blockers Gd(3+) (10 microM), La(3+) (10 microM), and Ni(2+) (100 microM) reduced CCE by 67% (n = 14), 65% (n = 11), and 55% (n = 9), respectively. These agents did not inhibit Ca(2+) entry when stores were not depleted. Conversely, CCE in IC was resistant to SKF-96365 (10 microM), wortmannin (10 microM), and nifedipine (1 microM). Spontaneous transient inward currents were recorded from IC voltage-clamped at -60 mV. These events were not significantly affected by Gd(3+) (10 microM) or La(3+) (10 microM) and were only slightly decreased in amplitude by 100 microM Ni(2+). The results from this study demonstrate that freshly dispersed IC from the rabbit urethra possess a CCE pathway. However, influx via this pathway does not appear to contribute to spontaneous activity in these cells.


Subject(s)
Biological Clocks/physiology , Calcium/metabolism , Myocytes, Smooth Muscle/physiology , Urethra/cytology , Urethra/physiology , Animals , Calcium Channel Blockers/pharmacology , Female , Gadolinium/pharmacology , Imidazoles/pharmacology , Lanthanum/pharmacology , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Nickel/pharmacology , Patch-Clamp Techniques , Rabbits
15.
Am J Physiol Heart Circ Physiol ; 284(6): H2007-14, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12742824

ABSTRACT

We examined the contributions of the cotransmitters norepinephrine (NE), ATP, and neuropeptide Y (NPY) to sympathetically evoked vasoconstriction in the rat tail artery in isolated vascular rings by using 1-100 stimulation impulses at 20 Hz. Phentolamine (2 microM), the alpha-adrenoceptor antagonist, markedly reduced responses to all stimuli, although responses to lower impulse numbers were reduced less than responses to longer trains. The purinergic receptor antagonist suramin (100 microM) reduced all responses, but to a much greater extent with few impulse trains. Responses were further reduced or abolished by addition of the second antagonist. Any remaining responses were abolished by the NPY-Y(1) receptor antagonist BIBP-3226 (75 nM). NPY had a direct agonist action and potentiated sympathetically mediated responses. NPY (75 nM) potentiated responses and BIBP-3226 decreased responses to 2- and 20-impulse trains. Both affected responses from 2 impulses to >20 impulses, but there was no preferential effect on purinergic contributions to responses because neurally released NPY potentiated both "pure" NE and ATP responses equally. We conclude that all three cotransmitters contribute significantly to vascular responses and their contribution varies markedly with impulse numbers. There is considerable synergy between cotransmitters, especially with lower impulse numbers where NPY contributions are greater than expected.


Subject(s)
Arginine/analogs & derivatives , Arteries/physiology , Neurotransmitter Agents/physiology , Sympathetic Nervous System/physiology , Tail/blood supply , Vasoconstriction/physiology , Adenosine Triphosphate/pharmacology , Adrenergic alpha-Antagonists/pharmacology , Animals , Arginine/pharmacology , Arteries/drug effects , Arteries/innervation , Electric Stimulation , In Vitro Techniques , Male , Muscle Contraction/drug effects , Neuropeptide Y/pharmacology , Norepinephrine/pharmacology , Purinergic P2 Receptor Antagonists , Rats , Rats, Sprague-Dawley , Regional Blood Flow/drug effects , Regional Blood Flow/physiology , Suramin/pharmacology , Sympathetic Nervous System/drug effects , Synaptic Transmission/drug effects , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...