Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Nat Commun ; 13(1): 830, 2022 02 11.
Article in English | MEDLINE | ID: mdl-35149693

ABSTRACT

Arrhythmias originating in scarred ventricular myocardium are a major cause of death, but the underlying mechanism allowing these rhythms to exist remains unknown. This gap in knowledge critically limits identification of at-risk patients and treatment once arrhythmias become manifest. Here we show that potassium voltage-gated channel subfamily E regulatory subunits 3 and 4 (KCNE3, KCNE4) are uniquely upregulated at arrhythmia sites within scarred myocardium. Ventricular arrhythmias occur in areas with a distinctive cardiomyocyte repolarization pattern, where myocyte tracts with short repolarization times connect to myocytes tracts with long repolarization times. We found this unique pattern of repolarization heterogeneity only in ventricular arrhythmia circuits. In contrast, conduction abnormalities were ubiquitous within scar. These repolarization heterogeneities are consistent with known functional effects of KCNE3 and KCNE4 on the slow delayed-rectifier potassium current. We observed repolarization heterogeneity using conventional cardiac electrophysiologic techniques that could potentially translate to identification of at-risk patients. The neutralization of the repolarization heterogeneities could represent a potential strategy for the elimination of ventricular arrhythmia circuits.


Subject(s)
Cicatrix/physiopathology , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Tachycardia, Ventricular/metabolism , Tachycardia, Ventricular/physiopathology , Animals , Arrhythmias, Cardiac/physiopathology , Electrocardiography , Electrophysiologic Techniques, Cardiac , Female , Guinea Pigs , Heart Ventricles/physiopathology , Humans , KCNQ1 Potassium Channel , Male , Myocardium/pathology , Potassium Channels, Voltage-Gated/metabolism
2.
Chonnam Med J ; 54(1): 63-71, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29399568

ABSTRACT

Purinergic receptors play an important role in regulating gastrointestinal (GI) motility. Interstitial cells of Cajal (ICCs) are pacemaker cells that regulate GI smooth muscle activity. We studied the functional roles of external adenosine 5'-triphosphate (ATP) on pacemaker activity in cultured ICCs from mouse small intestines by using the whole-cell patch clamp technique and intracellular Ca2+ ([Ca2+]i) imaging. External ATP dose-dependently depolarized the resting membrane and produced tonic inward pacemaker currents, and these effects were antagonized by suramin, a purinergic P2 receptor antagonist. ATP-induced effects on pacemaker currents were suppressed by an external Na+-free solution and inhibited by the nonselective cation channel blockers, flufenamic acid and niflumic acid. The removal of external Ca2+ or treatment with thapsigargin (inhibitor of Ca2+ uptake into endoplasmic reticulum) inhibited the ATP-induced effects on pacemaker currents. Spontaneous [Ca2+]i oscillations were enhanced by external ATP. These results suggest that external ATP modulates pacemaker activity by activating nonselective cation channels via external Ca2+ influx and [Ca2+]i release from the endoplasmic reticulum. Thus, it seems that activating the purinergic P2 receptor may modulate GI motility by acting on ICCs in the small intestine.

3.
Oncotarget ; 9(1): 21-36, 2018 Jan 02.
Article in English | MEDLINE | ID: mdl-29416593

ABSTRACT

Tumorigenesis results from the convergence of cell autonomous mutations and corresponding stromal changes that promote tumor cell growth. Senescent cells, which secrete a plethora of pro-tumorigenic factors termed the senescence-associated secretory phenotype (SASP), play an important role in tumor formation. Investigation into SASP regulation revealed that many but not all SASP factors are subject to NF-kB and p38MAPK regulation. However, many pro-tumorigenic SASP factors, including osteopontin (OPN), are not responsive to these canonical pathways leaving the regulation of these factors an open question. We report that the transcription factor c-Myb regulates OPN, IL-6, and IL-8 in addition to 57 other SASP factors. The regulation of OPN is direct as c-Myb binds to the OPN promoter in response to senescence. Further, OPN is also regulated by the known SASP regulator C/EBPß. In response to senescence, the full-length activating C/EBPß isoform LAP2 increases binding to the OPN, IL-6, and IL-8 promoters. The importance of both c-Myb and C/EBPß is underscored by our finding that the depletion of either factor reduces the ability of senescent fibroblasts to promote the growth of preneoplastic epithelial cells.

4.
Physiol Rep ; 5(14)2017 Jul.
Article in English | MEDLINE | ID: mdl-28754781

ABSTRACT

Estrogens have an important role in regulating detrusor smooth muscle (DSM) function. However, the underlying molecular and cellular mechanisms by which estrogens control human DSM excitability and contractility are not well known. Here, we used human DSM specimens from open bladder surgeries on 27 patients to elucidate the mechanism by which 17ß-estradiol regulates large conductance voltage- and Ca2+-activated K+ (BK) channels, the most prominent K+ channels in human DSM We employed single BK channel recordings on inside-out excised membrane patches, perforated whole-cell patch-clamp on freshly isolated DSM cells, and isometric tension recordings on DSM-isolated strips to investigate the mechanism by which 17ß-estradiol activates BK channels. 17ß-Estradiol (100 nmol/L) rapidly increased depolarization-induced whole-cell K+ currents in DSM cells. The 17ß-estradiol stimulatory effects on whole-cell BK currents were completely abolished by the selective BK channel inhibitor paxilline (1 µmol/L), clearly indicating that 17ß-estradiol specifically activates BK channels. 17ß-Estradiol also increased the frequency of ryanodine receptor-mediated transient BK currents. Single BK channel recordings showed that 17ß-estradiol (100 nmol/L) significantly increased the BK channel open probability of inside-out excised membrane patches, revealing that 17ß-estradiol activates BK channels directly. 17ß-Estradiol reduced spontaneous phasic contractions of human DSM-isolated strips in a concentration-dependent manner (100 nmol/L-1 µmol/L), and this effect was blocked by paxilline (1 µmol/L). 17ß-Estradiol (100 nmol/L) also reduced nerve-evoked contractions of human DSM-isolated strips. Collectively, our results reveal that 17ß-estradiol plays a critical role in regulating human DSM function through a direct nongenomic activation of BK channels.


Subject(s)
Estradiol/pharmacology , Estrogens/pharmacology , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Myocytes, Smooth Muscle/physiology , Action Potentials , Aged , Cells, Cultured , Female , Humans , Male , Muscle Contraction , Myocytes, Smooth Muscle/drug effects , Urinary Bladder/cytology
5.
J Biol Chem ; 292(37): 15216-15224, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28717002

ABSTRACT

Faithful DNA replication is essential for genome stability. To ensure accurate replication, numerous complex and redundant replication and repair mechanisms function in tandem with the core replication proteins to ensure DNA replication continues even when replication challenges are present that could impede progression of the replication fork. A unique topological challenge to the replication machinery is posed by RNA-DNA hybrids, commonly referred to as R-loops. Although R-loops play important roles in gene expression and recombination at immunoglobulin sites, their persistence is thought to interfere with DNA replication by slowing or impeding replication fork progression. Therefore, it is of interest to identify DNA-associated enzymes that help resolve replication-impeding R-loops. Here, using DNA fiber analysis, we demonstrate that human ribonuclease H1 (RNH1) plays an important role in replication fork movement in the mammalian nucleus by resolving R-loops. We found that RNH1 depletion results in accumulation of RNA-DNA hybrids, slowing of replication forks, and increased DNA damage. Our data uncovered a role for RNH1 in global DNA replication in the mammalian nucleus. Because accumulation of RNA-DNA hybrids is linked to various human cancers and neurodegenerative disorders, our study raises the possibility that replication fork progression might be impeded, adding to increased genomic instability and contributing to disease.


Subject(s)
DNA Replication , DNA/metabolism , RNA/metabolism , Replication Origin , Ribonuclease H/metabolism , Amino Acid Substitution , Chromosome Positioning , DNA/chemistry , DNA Damage , DNA Replication Timing , Gene Expression Regulation , Genomic Instability , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , In Situ Hybridization, Fluorescence , Mutation , Nucleic Acid Conformation , Nucleic Acid Hybridization , RNA/chemistry , RNA Interference , RNA, Messenger/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Ribonuclease H/antagonists & inhibitors , Ribonuclease H/genetics , Telomere Homeostasis
6.
Am J Physiol Renal Physiol ; 311(6): F1253-F1259, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27605581

ABSTRACT

In addition to improving sexual function, testosterone has been reported to have beneficial effects in ameliorating lower urinary tract symptoms by increasing bladder capacity and compliance, while decreasing bladder pressure. However, the cellular mechanisms by which testosterone regulates detrusor smooth muscle (DSM) excitability have not been elucidated. Here, we used amphotericin-B perforated whole cell patch-clamp and single channel recordings on inside-out excised membrane patches to investigate the regulatory role of testosterone in guinea pig DSM excitability. Testosterone (100 nM) significantly increased the depolarization-induced whole cell outward currents in DSM cells. The selective pharmacological inhibition of the large-conductance voltage- and Ca2+-activated K+ (BK) channels with paxilline (1 µM) completely abolished this stimulatory effect of testosterone, suggesting a mechanism involving BK channels. At a holding potential of -20 mV, DSM cells exhibited transient BK currents (TBKCs). Testosterone (100 nM) significantly increased TBKC activity in DSM cells. In current-clamp mode, testosterone (100 nM) significantly hyperpolarized the DSM cell resting membrane potential and increased spontaneous transient hyperpolarizations. Testosterone (100 nM) rapidly increased the single BK channel open probability in inside-out excised membrane patches from DSM cells, clearly suggesting a direct BK channel activation via a nongenomic mechanism. Live-cell Ca2+ imaging showed that testosterone (100 nM) caused a decrease in global intracellular Ca2+ concentration, consistent with testosterone-induced membrane hyperpolarization. In conclusion, the data provide compelling mechanistic evidence that under physiological conditions, testosterone at nanomolar concentrations directly activates BK channels in DSM cells, independent from genomic testosterone receptors, and thus regulates DSM excitability.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channels/metabolism , Membrane Potentials/drug effects , Myocytes, Smooth Muscle/drug effects , Signal Transduction/drug effects , Testosterone/pharmacology , Urinary Bladder/drug effects , Animals , Calcium/metabolism , Guinea Pigs , Male , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Myocytes, Smooth Muscle/metabolism , Patch-Clamp Techniques , Urinary Bladder/metabolism
7.
Channels (Austin) ; 10(5): 355-364, 2016 Sep 02.
Article in English | MEDLINE | ID: mdl-27101440

ABSTRACT

Contraction and relaxation of urinary bladder smooth muscle cells (UBSMCs) represent the important physiological functions of the bladder. Contractile responses in UBSMCs are regulated by a number of ion channels including big-conductance Ca2+- activated K+ (BK) channels. Great progress has been made in studies of BK channels in UBSMCs. The intent of this review is to summarize recent exciting findings with respect to the functional interactions of BK channels with muscarinic receptors, ryanodine receptors (RyRs) and inositol triphosphate receptors (IP3Rs) as well as their functional importance under normal and pathophysiological conditions. BK channels are highly expressed in UBSMCs. Activation of muscarinic M3 receptors inhibits the BK channel activity, facilitates opening of voltage-dependent Ca2+ (CaV) channels, and thereby enhances excitability and contractility of UBSMCs. Signaling molecules and regulatory mechanisms involving RyRs and IP3Rs have a significant effect on functions of BK channels and thereby regulate cellular responses in UBSMCs under normal and pathophysiological conditions including overactive bladders. Moreover, BK channels may represent a novel target for the treatment of bladder dysfunctions.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channels/physiology , Myocytes, Smooth Muscle/physiology , Urinary Bladder/physiology , Animals , Humans , Inositol 1,4,5-Trisphosphate Receptors/physiology , Receptors, Muscarinic/physiology , Ryanodine Receptor Calcium Release Channel/physiology , Urinary Bladder/physiopathology
8.
Am J Physiol Cell Physiol ; 310(7): C600-11, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26791488

ABSTRACT

Transient receptor potential melastatin 4 (TRPM4) channels are Ca(2+)-activated nonselective cation channels that have been recently identified as regulators of detrusor smooth muscle (DSM) function in rodents. However, their expression and function in human DSM remain unexplored. We provide insights into the functional role of TRPM4 channels in human DSM under physiological conditions. We used a multidisciplinary experimental approach, including RT-PCR, Western blotting, immunohistochemistry and immunocytochemistry, patch-clamp electrophysiology, and functional studies of DSM contractility. DSM samples were obtained from patients without preoperative overactive bladder symptoms. RT-PCR detected mRNA transcripts for TRPM4 channels in human DSM whole tissue and freshly isolated single cells. Western blotting and immunohistochemistry with confocal microscopy revealed TRPM4 protein expression in human DSM. Immunocytochemistry further detected TRPM4 protein expression in DSM single cells. Patch-clamp experiments showed that 9-phenanthrol, a selective TRPM4 channel inhibitor, significantly decreased the transient inward cation currents and voltage step-induced whole cell currents in freshly isolated human DSM cells. In current-clamp mode, 9-phenanthrol hyperpolarized the human DSM cell membrane potential. Furthermore, 9-phenanthrol attenuated the spontaneous phasic, carbachol-induced and nerve-evoked contractions in human DSM isolated strips. Significant species-related differences in TRPM4 channel activity between human, rat, and guinea pig DSM were revealed, suggesting a more prominent physiological role for the TRPM4 channel in the regulation of DSM function in humans than in rodents. In conclusion, TRPM4 channels regulate human DSM excitability and contractility and are critical determinants of human urinary bladder function. Thus, TRPM4 channels could represent promising novel targets for the pharmacological or genetic control of overactive bladder.


Subject(s)
Muscle Contraction/physiology , Muscle, Smooth/metabolism , TRPM Cation Channels/metabolism , Urinary Bladder/metabolism , Aged , Aged, 80 and over , Animals , Blotting, Western , Female , Guinea Pigs , Humans , Immunohistochemistry , Male , Microscopy, Confocal , Middle Aged , Patch-Clamp Techniques , Rats , Reverse Transcriptase Polymerase Chain Reaction , Species Specificity
9.
PLoS One ; 10(11): e0141950, 2015.
Article in English | MEDLINE | ID: mdl-26536038

ABSTRACT

Estrogen replacement therapies have been suggested to be beneficial in alleviating symptoms of overactive bladder. However, the precise regulatory mechanisms of estrogen in urinary bladder smooth muscle (UBSM) at the cellular level remain unknown. Large conductance voltage- and Ca2+-activated K+ (BK) channels, which are key regulators of UBSM function, are suggested to be non-genomic targets of estrogens. This study provides an electrophysiological investigation into the role of UBSM BK channels as direct targets for 17ß-estradiol, the principle estrogen in human circulation. Single BK channel recordings on inside-out excised membrane patches and perforated whole cell patch-clamp were applied in combination with the BK channel selective inhibitor paxilline to elucidate the mechanism of regulation of BK channel activity by 17ß-estradiol in freshly-isolated guinea pig UBSM cells. 17ß-Estradiol (100 nM) significantly increased the amplitude of depolarization-induced whole cell steady-state BK currents and the frequency of spontaneous transient BK currents in freshly-isolated UBSM cells. The increase in whole cell BK currents by 17ß-estradiol was eliminated upon blocking BK channels with paxilline. 17ß-Estradiol (100 nM) significantly increased (~3-fold) the single BK channel open probability, indicating direct 17ß-estradiol-BK channel interactions. 17ß-Estradiol (100 nM) caused a significant hyperpolarization of the membrane potential of UBSM cells, and this hyperpolarization was reversed by blocking the BK channels with paxilline. 17ß-Estradiol (100 nM) had no effects on L-type voltage-gated Ca2+ channel currents recorded under perforated patch-clamp conditions. This study reveals a new regulatory mechanism in the urinary bladder whereby BK channels are directly activated by 17ß-estradiol to reduce UBSM cell excitability.


Subject(s)
Estradiol/pharmacology , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Membrane Potentials/drug effects , Muscle, Smooth/metabolism , Urinary Bladder/cytology , Animals , Calcium Channels, L-Type/chemistry , Calcium Channels, L-Type/metabolism , Cells, Cultured , Guinea Pigs , Indoles/pharmacology , Large-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Male , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology
10.
J Biol Chem ; 290(24): 15133-45, 2015 Jun 12.
Article in English | MEDLINE | ID: mdl-25922071

ABSTRACT

The existence of redundant replication and repair systems that ensure genome stability underscores the importance of faithful DNA replication. Nowhere is this complexity more evident than in challenging DNA templates, including highly repetitive or transcribed sequences. Here, we demonstrate that flap endonuclease 1 (FEN1), a canonical lagging strand DNA replication protein, is required for normal, complete leading strand replication at telomeres. We find that the loss of FEN1 nuclease activity, but not DNA repair activities, results in leading strand-specific telomere fragility. Furthermore, we show that FEN1 depletion-induced telomere fragility is increased by RNA polymerase II inhibition and is rescued by ectopic RNase H1 expression. These data suggest that FEN1 limits leading strand-specific telomere fragility by processing RNA:DNA hybrid/flap intermediates that arise from co-directional collisions occurring between the replisome and RNA polymerase. Our data reveal the first molecular mechanism for leading strand-specific telomere fragility and the first known role for FEN1 in leading strand DNA replication. Because FEN1 mutations have been identified in human cancers, our findings raise the possibility that unresolved RNA:DNA hybrid structures contribute to the genomic instability associated with cancer.


Subject(s)
Flap Endonucleases/metabolism , Telomere , Blotting, Western , DNA Damage , DNA Replication , Flap Endonucleases/genetics , HEK293 Cells , Humans , In Situ Hybridization, Fluorescence , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
11.
Pflugers Arch ; 467(4): 665-75, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24867682

ABSTRACT

Activation of muscarinic acetylcholine receptors (mAChRs) constitutes the primary mechanism for enhancing excitability and contractility of human detrusor smooth muscle (DSM). Since the large-conductance Ca(2+)-activated K(+) (KCa1.1) channels are key regulators of human DSM function, we investigated whether mAChR activation increases human DSM excitability by inhibiting KCa1.1 channels. We used the mAChR agonist, carbachol, to determine the changes in KCa1.1 channel activity upon mAChR activation in freshly isolated human DSM cells obtained from open bladder surgeries using the perforated whole cell and single KCa1.1 channel patch-clamp recordings. Human DSM cells were collected from 29 patients (23 males and 6 females, average age of 65.9 ± 1.5 years). Carbachol inhibited the amplitude and frequency of KCa1.1 channel-mediated spontaneous transient outward currents and spontaneous transient hyperpolarizations, which are triggered by the release of Ca(2+) from ryanodine receptors. Carbachol also caused membrane potential depolarization, which was not observed in the presence of iberiotoxin, a KCa1.1 channel inhibitor, indicating the critical role of the KCa1.1 channels. The potential direct carbachol effects on KCa1.1 channels were examined under conditions of removing the major cellular Ca(2+) sources for KCa1.1 channel activation with pharmacological inhibitors (thapsigargin, ryanodine, and nifedipine). In the presence of these inhibitors, carbachol did not affect the single KCa1.1 channel open probability and mean KCa1.1 channel conductance (cell-attached configuration) or depolarization-induced whole cell steady-state KCa1.1 currents. The data support the concept that mAChR activation triggers indirect functional KCa1.1 channel inhibition mediated by intracellular Ca(2+), thus increasing the excitability in human DSM cells.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Myocytes, Smooth Muscle/metabolism , Receptors, Muscarinic/metabolism , Urinary Bladder/metabolism , Action Potentials , Aged , Calcium/metabolism , Carbachol/pharmacology , Cholinergic Agonists/pharmacology , Female , Humans , Male , Middle Aged , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/physiology , Peptides/pharmacology , Potassium Channel Blockers/pharmacology , Ryanodine Receptor Calcium Release Channel/metabolism , Urinary Bladder/cytology
12.
Eur J Pharmacol ; 738: 179-85, 2014 Sep 05.
Article in English | MEDLINE | ID: mdl-24886877

ABSTRACT

Prostaglandin E2 (PGE2) is an essential signaling molecule involved in the regulation of detrusor smooth muscle (DSM) function. However, the underlying regulatory mechanism by which PGE2 augments DSM cell excitability and contractility is not well understood. Here, we investigated whether PGE2 inhibits the large conductance voltage- and Ca(2+)-activated K(+) (BK) channels in guinea pig DSM, thereby increasing DSM excitability and contractility. We used a multidisciplinary experimental approach including amphotericin-B perforated patch-clamp electrophysiology and live-cell Ca(2+) imaging in native freshly-isolated DSM cells, isometric tension recordings of intact DSM strips, and pharmacological tools to investigate BK channel regulation by PGE2 in guinea pig DSM. PGE2 increased the spontaneous phasic contractions of isolated DSM strips in a concentration-dependent manner (10 nM-10 µM). BK channel inhibition with paxilline (1 µM) attenuated the PGE2-induced DSM phasic contractions, suggesting that BK channels are involved in the mechanism of PGE2-induced DSM contractions. PGE2 (10 µM) increased the intracellular Ca(2+) levels in freshly-isolated DSM cells. PGE2 (10 µM) also caused an inhibition of the amplitude and frequency of spontaneous transient BK currents in DSM cells. Moreover, PGE2 (10 µM) did not affect the amplitude of whole cell steady-state BK currents in DSM cells. Our findings provide strong experimental evidence that PGE2 leads to an inhibition of the spontaneous transient BK currents, elevation of intracellular Ca(2+) levels in freshly-isolated DSM cells, and augmentation of DSM phasic contractions. Thus, we have revealed a novel mechanism that BK channels mediate PGE2-induced contractions in guinea pig DSM.


Subject(s)
Dinoprostone/pharmacology , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , Potassium Channels, Calcium-Activated/metabolism , Urinary Bladder/drug effects , Urinary Bladder/physiology , Animals , Calcium/metabolism , Electrophysiological Phenomena/drug effects , Guinea Pigs , Intracellular Space/drug effects , Intracellular Space/metabolism , Muscle Contraction/drug effects , Muscle, Smooth/cytology , Muscle, Smooth/metabolism
13.
Am J Physiol Cell Physiol ; 306(5): C460-70, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24352333

ABSTRACT

Large-conductance voltage- and Ca(2+)-activated K(+) (BK) channels are critical regulators of detrusor smooth muscle (DSM) excitability and contractility. PKC modulates the contraction of DSM and BK channel activity in non-DSM cells; however, the cellular mechanism regulating the PKC-BK channel interaction in DSM remains unknown. We provide a novel mechanistic insight into BK channel regulation by PKC in DSM. We used patch-clamp electrophysiology, live-cell Ca(2+) imaging, and functional studies of DSM contractility to elucidate BK channel regulation by PKC at cellular and tissue levels. Voltage-clamp experiments showed that pharmacological activation of PKC with PMA inhibited the spontaneous transient BK currents in native freshly isolated guinea pig DSM cells. Current-clamp recordings revealed that PMA significantly depolarized DSM membrane potential and inhibited the spontaneous transient hyperpolarizations in DSM cells. The PMA inhibitory effects on DSM membrane potential were completely abolished by the selective BK channel inhibitor paxilline. Activation of PKC with PMA did not affect the amplitude of the voltage-step-induced whole cell steady-state BK current or the single BK channel open probability (recorded in cell-attached mode) upon inhibition of all major Ca(2+) sources for BK channel activation with thapsigargin, ryanodine, and nifedipine. PKC activation with PMA elevated intracellular Ca(2+) levels in DSM cells and increased spontaneous phasic and nerve-evoked contractions of DSM isolated strips. Our results support the concept that PKC activation leads to a reduction of BK channel activity in DSM via a Ca(2+)-dependent mechanism, thus increasing DSM contractility.


Subject(s)
Ion Channel Gating , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Muscle Contraction , Muscle, Smooth/enzymology , Potassium/metabolism , Protein Kinase C/metabolism , Urinary Bladder/enzymology , Animals , Calcium Signaling , Electric Stimulation , Enzyme Activation , Enzyme Activators/pharmacology , Evoked Potentials , Guinea Pigs , Large-Conductance Calcium-Activated Potassium Channels/drug effects , Male , Membrane Potentials , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/innervation , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Time Factors , Urinary Bladder/drug effects , Urinary Bladder/innervation
14.
Channels (Austin) ; 7(6): 537-40, 2013.
Article in English | MEDLINE | ID: mdl-24037125

ABSTRACT

The Ca (2+)-activated monovalent cation selective transient receptor potential melastatin 4 (TRPM4) channel has been recently identified in detrusor smooth muscle (DSM) of the urinary bladder. Two recent publications by our research group provide evidence in support of the novel hypothesis that TRPM4 channels enhance DSM excitability and contractility. This is a critical question as prior studies have primarily targeted hyperpolarizing currents facilitated by K(+) channels, but the depolarizing component in DSM cells is not well understood. For the first time, we utilized the selective TRPM4 channel inhibitor, 9-phenanthrol, to investigate TRPM4 channel functional effects in DSM at both cellular and tissue levels in rodents. Our new data presented here showed that in rat DSM cells, 9-phenanthrol attenuates spontaneous inward currents in the presence of the muscarinic receptor agonist, carbachol, thus reducing DSM cell excitability. In support of our original hypothesis, we found that TRPM4 channel mRNA levels are much higher in DSM vs. vascular smooth muscle and that inhibition of TRPM4 channels can potentially attenuate DSM excitability. Thus, we postulate the novel concept that selective pharmacological inhibition of TRPM4 channels can limit both excitability and contractility of DSM.


Subject(s)
Muscle, Smooth/physiology , Myocytes, Smooth Muscle/physiology , TRPM Cation Channels/physiology , Urinary Bladder/physiology , Animals , Male
15.
PLoS One ; 8(7): e68052, 2013.
Article in English | MEDLINE | ID: mdl-23861849

ABSTRACT

Patients suffering from a variety of neurological diseases such as spinal cord injury, Parkinson's disease, and multiple sclerosis often develop neurogenic detrusor overactivity (NDO), which currently lacks a universally effective therapy. Here, we tested the hypothesis that NDO is associated with changes in detrusor smooth muscle (DSM) large conductance Ca(2+)-activated K(+) (BK) channel expression and function. DSM tissue samples from 33 patients were obtained during open bladder surgeries. NDO patients were clinically characterized preoperatively with pressure-flow urodynamics demonstrating detrusor overactivity, in the setting of a clinically relevant neurological condition. Control patients did not have overactive bladder and did not have a clinically relevant neurological disease. We conducted quantitative polymerase chain reactions (qPCR), perforated patch-clamp electrophysiology on freshly-isolated DSM cells, and functional studies on DSM contractility. qPCR experiments revealed that DSM samples from NDO patients showed decreased BK channel mRNA expression in comparison to controls. Patch-clamp experiments demonstrated reduced whole cell and transient BK currents (TBKCs) in freshly-isolated DSM cells from NDO patients. Functional studies on DSM contractility showed that spontaneous phasic contractions had a decreased sensitivity to iberiotoxin, a selective BK channel inhibitor, in DSM strips isolated from NDO patients. These results reveal the novel finding that NDO is associated with decreased DSM BK channel expression and function leading to increased DSM excitability and contractility. BK channel openers or BK channel gene transfer could be an alternative strategy to control NDO. Future clinical trials are needed to evaluate the value of BK channel opening drugs or gene therapies for NDO treatment and to identify any possible adverse effects.


Subject(s)
Gene Expression Regulation , Genetic Association Studies , Large-Conductance Calcium-Activated Potassium Channels/genetics , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Urinary Bladder, Overactive/genetics , Urinary Bladder, Overactive/metabolism , Aged , Female , Humans , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/antagonists & inhibitors , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Large-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Male , Membrane Potentials/drug effects , Middle Aged , Muscle Contraction/drug effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Peptides/pharmacology , Urinary Bladder, Overactive/physiopathology
16.
Am J Physiol Cell Physiol ; 305(2): C207-14, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23703523

ABSTRACT

Large conductance voltage- and Ca(2+)-activated K(+) (BK) channels are key regulators of detrusor smooth muscle (DSM) contraction and relaxation during urine voiding and storage. Here, we explored whether BK channels are regulated by muscarinic receptors (M-Rs) in native freshly isolated rat DSM cells under physiological conditions using the perforated whole cell patch-clamp technique and pharmacological inhibitors. M-R activation with carbachol (1 µM) initially evoked large transient outward BK currents, followed by inhibition of the spontaneous transient outward BK currents (STBKCs) in DSM cells. Carbachol (1 µM) also inhibited the amplitude and frequency of spontaneous transient hyperpolarizations (STHs) and depolarized the DSM cell membrane potential. Selective inhibition of the muscarinic M3 receptors (M3-Rs) with 4-diphenylacetoxy-N-methylpiperidine (4-DAMP; 0.1 µM), but not muscarinic M2 receptors with methoctramine (1 µM), blocked the carbachol inhibitory effects on STBKCs. Furthermore, blocking the inositol 1,4,5-triphosphate (IP3) receptors with xestospongin-C (1 µM) inhibited the carbachol-induced large transient outward BK currents without affecting carbachol inhibitory effects on STBKCs. Upon pharmacological inhibition of all known cellular sources of Ca(2+) for BK channel activation, carbachol (1 µM) did not affect the voltage-step-induced steady-state BK currents, suggesting that the muscarinic effects in DSM cells are mediated by mobilization of intracellular Ca(2+). In conclusion, our findings provide strong evidence that activation of M3-Rs leads to inhibition of the STBKCs, STHs, and depolarization of DSM cells. Collectively, the data suggest the existence of functional interactions between BK channels and M3-Rs at a cellular level in DSM.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channels/metabolism , Myocytes, Smooth Muscle/metabolism , Receptor, Muscarinic M3/antagonists & inhibitors , Receptor, Muscarinic M3/metabolism , Urinary Bladder/cytology , Amphotericin B , Animals , Carbachol/pharmacology , Cholinergic Agonists/pharmacology , Diamines/pharmacology , Evoked Potentials/drug effects , Evoked Potentials/physiology , Gene Expression Regulation/physiology , Large-Conductance Calcium-Activated Potassium Channels/genetics , Male , Membrane Potentials , Muscarinic Antagonists/pharmacology , Patch-Clamp Techniques , Piperidines/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Muscarinic M3/genetics
17.
Proc Natl Acad Sci U S A ; 110(10): E878-87, 2013 Mar 05.
Article in English | MEDLINE | ID: mdl-23431150

ABSTRACT

Although the ribosome is a very general catalyst, it cannot synthesize all protein sequences equally well. For example, ribosomes stall on the secretion monitor (SecM) leader peptide to regulate expression of a downstream gene. Using a genetic selection in Escherichia coli, we identified additional nascent peptide motifs that stall ribosomes. Kinetic studies show that some nascent peptides dramatically inhibit rates of peptide release by release factors. We find that residues upstream of the minimal stalling motif can either enhance or suppress this effect. In other stalling motifs, peptidyl transfer to certain aminoacyl-tRNAs is inhibited. In particular, three consecutive Pro codons pose a challenge for elongating ribosomes. The translation factor elongation factor P, which alleviates pausing at polyproline sequences, has little or no effect on other stalling peptides. The motifs that we identified are underrepresented in bacterial proteomes and show evidence of stalling on endogenous E. coli proteins.


Subject(s)
Escherichia coli Proteins/biosynthesis , Escherichia coli/genetics , Escherichia coli/metabolism , Amino Acid Motifs , Amino Acid Sequence , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Genes, Reporter , Models, Biological , Molecular Sequence Data , Peptide Chain Elongation, Translational , Peptide Chain Termination, Translational , Peptide Elongation Factors/metabolism , Peptides/chemistry , Peptides/genetics , Peptides/metabolism , Protein Synthesis Inhibitors/metabolism , Ribosomes/metabolism , Two-Hybrid System Techniques
18.
Am J Physiol Renal Physiol ; 304(7): F918-29, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23283997

ABSTRACT

The TRPM4 channel is a Ca(2+)-activated, monovalent cation-selective channel of the melastatin transient receptor potential (TRPM) family. The TRPM4 channel is implicated in the regulation of many cellular processes including the immune response, insulin secretion, and pressure-induced vasoconstriction of cerebral arteries. However, the expression and function of the TRPM4 channels in detrusor smooth muscle (DSM) have not yet been explored. Here, we provide the first molecular, electrophysiological, and functional evidence for the presence of TRPM4 channels in rat DSM. We detected the expression of TRPM4 channels at mRNA and protein levels in freshly isolated DSM single cells and DSM tissue using RT-PCR, Western blotting, immunohistochemistry, and immunocytochemistry. 9-Hydroxyphenanthrene (9-phenanthrol), a novel selective inhibitor of TRPM4 channels, was used to examine their role in DSM function. In perforated patch-clamp recordings using freshly isolated rat DSM cells, 9-phenanthrol (30 µM) decreased the spontaneous inward current activity at -70 mV. Real-time DSM live-cell Ca(2+) imaging showed that selective inhibition of TRPM4 channels with 9-phenanthrol (30 µM) significantly reduced the intracellular Ca(2+) levels. Isometric DSM tension recordings revealed that 9-phenanthrol (0.1-30 µM) significantly inhibited the amplitude, muscle force integral, and frequency of the spontaneous phasic and pharmacologically induced contractions of rat DSM isolated strips. 9-Phenanthrol also decreased the amplitude and muscle force integral of electrical field stimulation-induced contractions. In conclusion, this is the first study to examine the expression and provide evidence for TRPM4 channels as critical regulators of rat DSM excitability and contractility.


Subject(s)
Muscle, Smooth/physiology , TRPM Cation Channels/physiology , Urinary Bladder/physiology , Animals , Male , Muscle Contraction/drug effects , Patch-Clamp Techniques , Phenanthrenes/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , TRPM Cation Channels/biosynthesis , Urinary Bladder/drug effects
19.
Am J Physiol Cell Physiol ; 304(5): C467-77, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23302778

ABSTRACT

Members of the transient receptor potential (TRP) channel superfamily, including the Ca(2+)-activated monovalent cation-selective TRP melastatin 4 (TRPM4) channel, have been recently identified in the urinary bladder. However, their expression and function at the level of detrusor smooth muscle (DSM) remain largely unexplored. In this study, for the first time we investigated the role of TRPM4 channels in guinea pig DSM excitation-contraction coupling using a multidisciplinary approach encompassing protein detection, electrophysiology, live-cell Ca(2+) imaging, DSM contractility, and 9-phenanthrol, a recently characterized selective inhibitor of the TRPM4 channel. Western blot and immunocytochemistry experiments demonstrated the expression of the TRPM4 channel in whole DSM tissue and freshly isolated DSM cells with specific localization on the plasma membrane. Perforated whole cell patch-clamp recordings and real-time Ca(2+) imaging experiments with fura 2-AM, both using freshly isolated DSM cells, revealed that 9-phenanthrol (30 µM) significantly reduced the cation current and decreased intracellular Ca(2+) levels. 9-Phenanthrol (0.1-30 µM) significantly inhibited spontaneous, 0.1 µM carbachol-induced, 20 mM KCl-induced, and nerve-evoked contractions in guinea pig DSM-isolated strips with IC50 values of 1-7 µM and 70-80% maximum inhibition. 9-Phenanthrol also reduced nerve-evoked contraction amplitude induced by continuous repetitive electrical field stimulation of 10-Hz frequency and shifted the frequency-response curve (0.5-50 Hz) relative to the control. Collectively, our data demonstrate the novel finding that TRPM4 channels are expressed in guinea pig DSM and reveal their critical role in the regulation of guinea pig DSM excitation-contraction coupling.


Subject(s)
Muscle, Smooth/physiology , Myocytes, Smooth Muscle/physiology , TRPM Cation Channels/physiology , Animals , Calcium/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Guinea Pigs , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Patch-Clamp Techniques/methods , Phenanthrenes/pharmacology , TRPM Cation Channels/metabolism
20.
Br J Pharmacol ; 168(7): 1611-25, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23145946

ABSTRACT

BACKGROUND AND PURPOSE: Overactive bladder (OAB) is often associated with abnormally increased detrusor smooth muscle (DSM) contractions. We used NS309, a selective and potent opener of the small or intermediate conductance Ca(2+) -activated K(+) (SK or IK, respectively) channels, to evaluate how SK/IK channel activation modulates DSM function. EXPERIMENTAL APPROACH: We employed single-cell RT-PCR, immunocytochemistry, whole cell patch-clamp in freshly isolated rat DSM cells and isometric tension recordings of isolated DSM strips to explore how the pharmacological activation of SK/IK channels with NS309 modulates DSM function. KEY RESULTS: We detected SK3 but not SK1, SK2 or IK channels expression at both mRNA and protein levels by RT-PCR and immunocytochemistry in DSM single cells. NS309 (10 µM) significantly increased the whole cell SK currents and hyperpolarized DSM cell resting membrane potential. The NS309 hyperpolarizing effect was blocked by apamin, a selective SK channel inhibitor. NS309 inhibited the spontaneous phasic contraction amplitude, force, frequency, duration and tone of isolated DSM strips in a concentration-dependent manner. The inhibitory effect of NS309 on spontaneous phasic contractions was blocked by apamin but not by TRAM-34, indicating no functional role of the IK channels in rat DSM. NS309 also significantly inhibited the pharmacologically and electrical field stimulation-induced DSM contractions. CONCLUSIONS AND IMPLICATIONS: Our data reveal that SK3 channel is the main SK/IK subtype in rat DSM. Pharmacological activation of SK3 channels with NS309 decreases rat DSM cell excitability and contractility, suggesting that SK3 channels might be potential therapeutic targets to control OAB associated with detrusor overactivity.


Subject(s)
Indoles/pharmacology , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Muscle, Smooth/drug effects , Oximes/pharmacology , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Urinary Bladder/drug effects , Animals , Apamin/pharmacology , Cell Size/drug effects , Electric Stimulation , Female , In Vitro Techniques , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Ion Channel Gating , Male , Membrane Potentials/drug effects , Muscle Contraction/drug effects , Muscle, Smooth/physiology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/physiology , Pyrazoles/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Small-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Small-Conductance Calcium-Activated Potassium Channels/genetics , Urinary Bladder/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...