Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 26
1.
Cell Mol Neurobiol ; 43(3): 1105-1127, 2023 Apr.
Article En | MEDLINE | ID: mdl-35695980

The striatum is especially vulnerable to HIV-1 infection, with medium spiny neurons (MSNs) exhibiting marked synaptodendritic damage that can be exacerbated by opioid use disorder. Despite known structural defects in MSNs co-exposed to HIV-1 Tat and opioids, the pathophysiological sequelae of sustained HIV-1 exposure and acute comorbid effects of opioids on dopamine D1 and D2 receptor-expressing (D1 and D2) MSNs are unknown. To address this question, Drd1-tdTomato- or Drd2-eGFP-expressing reporter and conditional HIV-1 Tat transgenic mice were interbred. MSNs in ex vivo slices from male mice were assessed by whole-cell patch-clamp electrophysiology and filled with biocytin to explore the functional and structural effects of progressive Tat and acute morphine exposure. Although the excitability of both D1 and D2 MSNs increased following 48 h of Tat exposure, D1 MSN firing rates decreased below control (Tat-) levels following 2 weeks and 1 month of Tat exposure but returned to control levels after 2 months. D2 neurons continued to display Tat-dependent increases in excitability at 2 weeks, but also returned to control levels following 1 and 2 months of Tat induction. Acute morphine exposure increased D1 MSN excitability irrespective of the duration of Tat exposure, while D2 MSNs were variably affected. That D1 and D2 MSN excitability would return to control levels was unexpected since both subpopulations displayed significant synaptodendritic degeneration and pathologic phospho-tau-Thr205 accumulation following 2 months of Tat induction. Thus, despite frank morphologic damage, D1 and D2 MSNs uniquely adapt to sustained Tat and acute morphine insults.


Dopamine , HIV-1 , Animals , Male , Mice , Analgesics, Opioid/pharmacology , Corpus Striatum/pathology , HIV-1/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Morphine/pharmacology , Neurons/metabolism , Receptors, Dopamine D1/metabolism
2.
Curr Top Membr ; 90: 37-63, 2022.
Article En | MEDLINE | ID: mdl-36368874

Lysosomal acid ceramidase (AC) has been reported to determine multivesicular body (MVB) fate and exosome secretion in different mammalian cells including coronary arterial endothelial cells (CAECs). However, this AC-mediated regulation of exosome release from CAECs and associated underlying mechanism remain poorly understood. In the present study, we hypothesized that AC controls lysosomal Ca2+ release through TRPML1 channel to regulate exosome release in murine CAECs. To test this hypothesis, we isolated and cultured CAECs from WT/WT and endothelial cell-specific Asah1 gene (gene encoding AC) knockout mice. Using these CAECs, we first demonstrated a remarkable increase in exosome secretion and significant reduction of lysosome-MVB interaction in CAECs lacking Asah1 gene compared to those cells from WT/WT mice. ML-SA1, a TRPML1 channel agonist, was found to enhance lysosome trafficking and increase lysosome-MVB interaction in WT/WT CAECs, but not in CAECs lacking Asah1 gene. However, sphingosine, an AC-derived sphingolipid, was able to increase lysosome movement and lysosome-MVB interaction in CAECs lacking Asah1 gene, leading to reduced exosome release from these cells. Moreover, Asah1 gene deletion was shown to substantially inhibit lysosomal Ca2+ release through suppression of TRPML1 channel activity in CAECs. Sphingosine as an AC product rescued the function of TRPML1 channel in CAECs lacking Asah1 gene. These results suggest that Asah1 gene defect and associated deficiency of AC activity may inhibit TRPML1 channel activity, thereby reducing MVB degradation by lysosome and increasing exosome release from CAECs. This enhanced exosome release from CAECs may contribute to the development of coronary arterial disease under pathological conditions.


Exosomes , Transient Receptor Potential Channels , Mice , Animals , Acid Ceramidase/genetics , Acid Ceramidase/metabolism , Exosomes/metabolism , Endothelial Cells/metabolism , Transient Receptor Potential Channels/metabolism , Sphingosine/metabolism , Lysosomes/metabolism , Mice, Knockout , Mammals/metabolism
3.
Am J Physiol Cell Physiol ; 322(3): C395-C409, 2022 03 01.
Article En | MEDLINE | ID: mdl-35080921

Dynamic chloride (Cl-) regulation is critical for synaptic inhibition. In mature neurons, Cl- influx and extrusion are primarily controlled by ligand-gated anion channels (GABAA and glycine receptors) and the potassium chloride cotransporter K+-Cl- cotransporter 2 (KCC2), respectively. Here, we report for the first time, to our knowledge, a presence of a new source of Cl- influx in striatal neurons with properties similar to chloride voltage-gated channel 1 (ClC-1). Using whole cell patch-clamp recordings, we detected an outwardly rectifying voltage-dependent current that was impermeable to the large anion methanesulfonate (MsO-). The anionic current was sensitive to the ClC-1 inhibitor 9-anthracenecarboxylic acid (9-AC) and the nonspecific blocker phloretin. The mean fractions of anionic current inhibition by MsO-, 9-AC, and phloretin were not significantly different, indicating that anionic current was caused by active ClC-1-like channels. In addition, we found that Cl- current was not sensitive to the transmembrane protein 16A (TMEM16A; Ano1) inhibitor Ani9 and that the outward Cl- rectification was preserved even at a very high intracellular Ca2+ concentration (2 mM), indicating that TMEM16B (Ano2) did not contribute to the total current. Western blotting and immunohistochemical analyses confirmed the presence of ClC-1 channels in the striatum mainly localized to the somata of striatal neurons. Finally, we found that 9-AC decreased action potential firing frequencies and increased excitability in medium spiny neurons (MSNs) expressing dopamine type 1 (D1) and type 2 (D2) receptors in the brain slices, respectively. We conclude that ClC-1-like channels are preferentially located at the somata of MSNs, are functional, and can modulate neuronal excitability.


Chlorides , Corpus Striatum , Chloride Channels/metabolism , Chlorides/metabolism , Corpus Striatum/metabolism , Neurons/metabolism , Patch-Clamp Techniques , Phloretin/metabolism , Phloretin/pharmacology , Receptors, Dopamine D2/metabolism
4.
eNeuro ; 8(3)2021.
Article En | MEDLINE | ID: mdl-33782102

About half the people infected with human immunodeficiency virus (HIV) have neurocognitive deficits that often include memory impairment and hippocampal deficits, which can be exacerbated by opioid abuse. To explore the effects of opioids and HIV on hippocampal CA1 pyramidal neuron structure and function, we induced HIV-1 transactivator of transcription (Tat) expression in transgenic mice for 14 d and co-administered time-release morphine or vehicle subcutaneous implants during the final 5 d (days 9-14) to establish steady-state morphine levels. Morphine was withheld from some ex vivo slices during recordings to begin to assess the initial pharmacokinetic consequences of opioid withdrawal. Tat expression reduced hippocampal CA1 pyramidal neuronal excitability at lower stimulating currents. Pyramidal cell firing rates were unaffected by continuous morphine exposure. Behaviorally, exposure to Tat or high dosages of morphine impaired spatial memory Exposure to Tat and steady-state levels of morphine appeared to have largely independent effects on pyramidal neuron structure and function, a response that is distinct from other vulnerable brain regions such as the striatum. By contrast, acutely withholding morphine (from morphine-tolerant ex vivo slices) revealed unique and selective neuroadaptive shifts in CA1 pyramidal neuronal excitability and dendritic plasticity, including some interactions with Tat. Collectively, the results show that opioid-HIV interactions in hippocampal area CA1 are more nuanced than previously assumed, and appear to vary depending on the outcome assessed and on the pharmacokinetics of morphine exposure.


HIV-1 , CA1 Region, Hippocampal/metabolism , HIV-1/metabolism , Hippocampus/metabolism , Morphine/pharmacology , Pyramidal Cells/metabolism , Spatial Learning , Trans-Activators , tat Gene Products, Human Immunodeficiency Virus/metabolism
5.
Am J Physiol Cell Physiol ; 318(2): C406-C421, 2020 02 01.
Article En | MEDLINE | ID: mdl-31851526

Nonselective cation channels, consistent with transient receptor potential melastatin-4 (TRPM4), regulate detrusor smooth muscle (DSM) function. TRPM4 channels can exist as homomers or assemble with sulfonylurea receptors (SURs) as complexes. We evaluated contributions of TRPM4/SUR-TRPM4 channels to DSM excitability and contractility by examining the effects of TRPM4/SUR-TRPM4 channel modulators 9-phenanthrol, glibenclamide, and diazoxide on freshly-isolated guinea pig DSM cells (amphotericin-B perforated patch-clamp electrophysiology) and mucosa-free DSM strips (isometric tension recordings). In DSM cells, complete removal of extracellular Na+ decreased voltage-step-induced cation (non-K+ selective) currents. At high positive membrane potentials, 9-phenanthrol at 100 µM attenuated voltage step-induced currents more effectively than at 30 µM, revealing concentration-dependent, voltage-sensitive inhibition. In comparison to 9-phenanthrol, glibenclamide (100 µM) displayed lower inhibition of cation currents. In the presence of glibenclamide (100 µM), 9-phenanthrol (100 µM) further decreased the currents. The SUR-TRPM4 complex activator diazoxide (100-300 µM) weakly inhibited the currents. 9-Phenanthrol, but not glibenclamide or diazoxide, increased cell capacitance (a cell surface area indicator). In contractility studies, glibenclamide displayed lower potencies than 9-phenanthrol attenuating spontaneous and 20 mM KCl-induced DSM phasic contractions. While both compounds showed similar maximum inhibitions on DSM spontaneous phasic contractions, glibenclamide was generally less efficacious on 20 mM KCl-induced phasic contractions. In summary, the observed differential effects of 9-phenanthrol and glibenclamide on DSM excitability and contractility support unique mechanisms for the two compounds. The data suggest that SUR-TRPM4 complexes do not contribute to DSM function. This study advances our understanding of pharmacological effects of glibenclamide and 9-phenanthrol on DSM cell cation currents.


Cations/metabolism , Glyburide/pharmacology , Muscle, Smooth/drug effects , Phenanthrenes/pharmacology , TRPM Cation Channels/antagonists & inhibitors , Urinary Bladder/drug effects , Urinary Bladder/metabolism , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Guinea Pigs , Male , Membrane Potentials/drug effects , Muscle Contraction/drug effects , Muscle, Smooth/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Patch-Clamp Techniques/methods
6.
Am J Physiol Cell Physiol ; 317(6): C1268-C1277, 2019 12 01.
Article En | MEDLINE | ID: mdl-31577513

Cl- channels serve as key regulators of excitability and contractility in vascular, intestinal, and airway smooth muscle cells. We recently reported a Cl- conductance in detrusor smooth muscle (DSM) cells. Here, we used the whole cell patch-clamp technique to further characterize biophysical properties and physiological regulators of the Cl- current in freshly isolated guinea pig DSM cells. The Cl- current demonstrated outward rectification arising from voltage-dependent gating of Cl- channels rather than the Cl- transmembrane gradient. An exposure of DSM cells to hypotonic extracellular solution (Δ 165 mOsm challenge) did not increase the Cl- current providing strong evidence that volume-regulated anion channels do not contribute to the Cl- current in DSM cells. The Cl- current was monotonically dependent on extracellular pH, larger and lower in magnitude at acidic (5.0) and basic pH (8.5) values, respectively. Additionally, intracellularly applied phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2] analog [PI(4,5)P2-diC8] increased the average Cl- current density by approximately threefold in a voltage-independent manner. The magnitude of the DSM whole cell Cl- current did not depend on the cell surface area (cell capacitance) regardless of the presence or absence of PI(4,5)P2-diC8, an intriguing finding that underscores the complex nature of Cl- channel expression and function in DSM cells. Removal of both extracellular Ca2+ and Mg2+ did not affect the DSM whole cell Cl- current, whereas Gd3+ (1 mM) potentiated the current. Collectively, our recent and present findings strongly suggest that Cl- channels are critical regulators of DSM excitability and are regulated by extracellular pH, Gd3+, and PI(4,5)P2.


Chloride Channels/physiology , Chlorides/metabolism , Gadolinium/metabolism , Membrane Potentials/physiology , Myocytes, Smooth Muscle/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Animals , Calcium/metabolism , Cations, Divalent , Chloride Channels/drug effects , Gadolinium/pharmacology , Guinea Pigs , Hydrogen-Ion Concentration , Ion Transport , Magnesium/metabolism , Male , Membrane Potentials/drug effects , Muscle Contraction/physiology , Muscle, Smooth/cytology , Muscle, Smooth/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Patch-Clamp Techniques , Phosphatidylinositol 4,5-Diphosphate/analogs & derivatives , Phosphatidylinositol 4,5-Diphosphate/pharmacology , Primary Cell Culture , Urinary Bladder/cytology , Urinary Bladder/metabolism
7.
Am J Physiol Cell Physiol ; 316(5): C698-C710, 2019 05 01.
Article En | MEDLINE | ID: mdl-30566392

Multiple types of Cl- channels regulate smooth muscle excitability and contractility in vascular, gastrointestinal, and airway smooth muscle cells. However, little is known about Cl- channels in detrusor smooth muscle (DSM) cells. Here, we used inside-out single channel and whole cell patch-clamp recordings for detailed biophysical and pharmacological characterizations of Cl- channels in freshly isolated guinea pig DSM cells. The recorded single Cl- channels displayed unique gating with multiple subconductive states, a fully opened single-channel conductance of 164 pS, and a reversal potential of -41.5 mV, which is close to the ECl of -65 mV, confirming preferential permeability to Cl-. The Cl- channel demonstrated strong voltage dependence of activation (half-maximum of mean open probability, V0.5, ~-20 mV) and robust prolonged openings at depolarizing voltages. The channel displayed similar gating when exposed intracellularly to solutions containing Ca2+-free or 1 mM Ca2+. In whole cell patch-clamp recordings, macroscopic current demonstrated outward rectification, inhibitions by 4,4'-diisothiocyano-2,2'-stilbenedisulfonic acid (DIDS) and niflumic acid, and insensitivity to chlorotoxin. The outward current was reversibly reduced by 94% replacement of extracellular Cl- with I-, Br-, or methanesulfonate (MsO-), resulting in anionic permeability sequence: Cl->Br->I->MsO-. While intracellular Ca2+ levels (0, 300 nM, and 1 mM) did not affect the amplitude of Cl- current and outward rectification, high Ca2+ slowed voltage-step current activation at depolarizing voltages. In conclusion, our data reveal for the first time the presence of a Ca2+-independent DIDS and niflumic acid-sensitive, voltage-dependent Cl- channel in the plasma membrane of DSM cells. This channel may be a key regulator of DSM excitability.


Cell Membrane/metabolism , Chloride Channels/metabolism , Myocytes, Smooth Muscle/metabolism , Urinary Bladder/cytology , Urinary Bladder/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cell Membrane/drug effects , Cells, Cultured , Chloride Channels/antagonists & inhibitors , Guinea Pigs , Male , Myocytes, Smooth Muscle/drug effects , Niflumic Acid/pharmacology , Urinary Bladder/drug effects
8.
Proc Natl Acad Sci U S A ; 114(2): E228-E236, 2017 01 10.
Article En | MEDLINE | ID: mdl-28003463

Skeletal muscle contractions are initiated by an increase in Ca2+ released during excitation-contraction (EC) coupling, and defects in EC coupling are associated with human myopathies. EC coupling requires communication between voltage-sensing dihydropyridine receptors (DHPRs) in transverse tubule membrane and Ca2+ release channel ryanodine receptor 1 (RyR1) in the sarcoplasmic reticulum (SR). Stac3 protein (SH3 and cysteine-rich domain 3) is an essential component of the EC coupling apparatus and a mutation in human STAC3 causes the debilitating Native American myopathy (NAM), but the nature of how Stac3 acts on the DHPR and/or RyR1 is unknown. Using electron microscopy, electrophysiology, and dynamic imaging of zebrafish muscle fibers, we find significantly reduced DHPR levels, functionality, and stability in stac3 mutants. Furthermore, stac3NAM myofibers exhibited increased caffeine-induced Ca2+ release across a wide range of concentrations in the absence of altered caffeine sensitivity as well as increased Ca2+ in internal stores, which is consistent with increased SR luminal Ca2+ These findings define critical roles for Stac3 in EC coupling and human disease.


Adaptor Proteins, Signal Transducing/physiology , Calcium Channels, L-Type/physiology , Muscle Fibers, Skeletal/physiology , Ryanodine Receptor Calcium Release Channel/physiology , Zebrafish Proteins/physiology , Adaptor Proteins, Signal Transducing/genetics , Animals , Animals, Genetically Modified , Caffeine/pharmacology , Calcium , Embryo, Nonmammalian , Microscopy, Electron , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/ultrastructure , Mutation , Myotonia Congenita , Zebrafish , Zebrafish Proteins/genetics
9.
Skelet Muscle ; 5: 4, 2015.
Article En | MEDLINE | ID: mdl-25717360

BACKGROUND: Ca(2+) influx through CaV1.1 is not required for skeletal muscle excitation-contraction coupling, but whether Ca(2+) permeation through CaV1.1 during sustained muscle activity plays a functional role in mammalian skeletal muscle has not been assessed. METHODS: We generated a mouse with a Ca(2+) binding and/or permeation defect in the voltage-dependent Ca(2+) channel, CaV1.1, and used Ca(2+) imaging, western blotting, immunohistochemistry, proximity ligation assays, SUnSET analysis of protein synthesis, and Ca(2+) imaging techniques to define pathways modulated by Ca(2+) binding and/or permeation of CaV1.1. We also assessed fiber type distributions, cross-sectional area, and force frequency and fatigue in isolated muscles. RESULTS: Using mice with a pore mutation in CaV1.1 required for Ca(2+) binding and/or permeation (E1014K, EK), we demonstrate that CaV1.1 opening is coupled to CaMKII activation and refilling of sarcoplasmic reticulum Ca(2+) stores during sustained activity. Decreases in these Ca(2+)-dependent enzyme activities alter downstream signaling pathways (Ras/Erk/mTORC1) that lead to decreased muscle protein synthesis. The physiological consequences of the permeation and/or Ca(2+) binding defect in CaV1.1 are increased fatigue, decreased fiber size, and increased Type IIb fibers. CONCLUSIONS: While not essential for excitation-contraction coupling, Ca(2+) binding and/or permeation via the CaV1.1 pore plays an important modulatory role in muscle performance.

11.
Biophys J ; 107(9): 2027-36, 2014 Nov 04.
Article En | MEDLINE | ID: mdl-25418088

Nuclear ion channels remain among the least studied and biophysically characterized channels. Although considerable progress has been made in characterizing calcium release channels in the nuclear membrane, very little is known regarding the properties of nuclear monovalent cationic channels. Here, we describe a method to isolate nuclei from adult skeletal muscle fibers that are suitable for electrophysiological experiments. Using this approach, we show for the first time, to our knowledge, that a nuclear monovalent cationic channel (NMCC) is prominently expressed in the inner membrane of nuclei isolated from flexor digitorum brevis skeletal muscle fibers of adult mice. In isotonic 140 mM KCl, the skeletal muscle NMCC exhibits a unitary conductance of ?160 pS and high, voltage-independent open probability. Based on single-channel reversal potential measurements, NMCCs are slightly more permeable to potassium ions over sodium (PK/PNa = 2.68 ± 0.21) and cesium (PK/PCs = 1.39 ± 0.03) ions. In addition, NMCCs do not permeate divalent cations, are inhibited by calcium ions, and demonstrate weak rectification in asymmetric Ca(2+)-containing solutions. Together, these studies characterize a voltage-independent NMCC in skeletal muscle, the properties of which are ideally suited to serve as a countercurrent mechanism during calcium release from the nuclear envelope.


Ion Channels/metabolism , Muscle Fibers, Skeletal/metabolism , Nuclear Envelope/metabolism , Animals , Calcium/metabolism , Cesium/metabolism , Fluorescent Antibody Technique , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Ions/metabolism , Mice, Inbred C57BL , Microscopy, Confocal , Patch-Clamp Techniques , Potassium/metabolism , Sodium/metabolism
12.
PLoS One ; 8(10): e77633, 2013.
Article En | MEDLINE | ID: mdl-24143248

Store-operated calcium entry (SOCE) channels play an important role in Ca(2+) signaling. Recently, excessive SOCE was proposed to play a central role in the pathogenesis of malignant hyperthermia (MH), a pharmacogenic disorder of skeletal muscle. We tested this hypothesis by characterizing SOCE current (ISkCRAC) magnitude, voltage dependence, and rate of activation in myotubes derived from two mouse models of anesthetic- and heat-induced sudden death: 1) type 1 ryanodine receptor (RyR1) knock-in mice (Y524S/+) and 2) calsequestrin 1 and 2 double knock-out (dCasq-null) mice. ISkCRAC voltage dependence and magnitude at -80 mV were not significantly different in myotubes derived from wild type (WT), Y524S/+ and dCasq-null mice. However, the rate of ISkCRAC activation upon repetitive depolarization was significantly faster at room temperature in myotubes from Y524S/+ and dCasq-null mice. In addition, the maximum rate of ISkCRAC activation in dCasq-null myotubes was also faster than WT at more physiological temperatures (35-37°C). Azumolene (50 µM), a more water-soluble analog of dantrolene that is used to reverse MH crises, failed to alter ISkCRAC density or rate of activation. Together, these results indicate that while an increased rate of ISkCRAC activation is a common characteristic of myotubes derived from Y524S/+ and dCasq-null mice and that the protective effects of azumolene are not due to a direct inhibition of SOCE channels.


Acceleration , Anesthetics/adverse effects , Calcium/metabolism , Death, Sudden/etiology , Electrophysiological Phenomena/drug effects , Hot Temperature/adverse effects , Muscle Fibers, Skeletal/metabolism , Animals , Calcium Channels/metabolism , Calcium-Binding Proteins/deficiency , Calcium-Binding Proteins/genetics , Calsequestrin/deficiency , Calsequestrin/genetics , Disease Models, Animal , Gene Knock-In Techniques , Gene Knockout Techniques , Imidazoles/adverse effects , Mice , Muscle Fibers, Skeletal/drug effects , Oxazoles/adverse effects , Ryanodine Receptor Calcium Release Channel/genetics
13.
Biophys J ; 103(2): 202-11, 2012 Jul 18.
Article En | MEDLINE | ID: mdl-22853897

Store-operated calcium entry (SOCE) is an important Ca(2+) entry pathway in skeletal muscle. However, direct electrophysiological recording and full characterization of the underlying SOCE current in skeletal muscle cells (I(SkCRAC)) has not been reported. Here, we characterized the biophysical properties, pharmacological profile, and molecular identity of I(SkCRAC) in skeletal myotubes, as well as the regulation of its rate of activation by temperature and the type I ryanodine receptor (RyR1). I(SkCRAC) exhibited many hallmarks of Ca(2+) release activated Ca(2+) currents (I(CRAC)): store dependence, strong inward rectification, positive reversal potential, limited cesium permeability, and sensitivity to SOCE channel blockers. I(SkCRAC) was reduced by siRNA knockdown of stromal interaction molecule 1 and expression of dominant negative Orai1. Average I(SkCRAC) current density at -80mV was 1.00 ± 0.05 pA/pF. In the presence of 20 mM intracellular EGTA, I(SkCRAC) activation occurred over tens of seconds during repetitive depolarization at 0.5Hz and was inhibited by treatment with 100 µM ryanodine. The rate of SOCE activation was reduced threefold in myotubes from RyR1-null mice and increased 4.6-fold at physiological temperatures (35-37°C). These results show that I(SkCRAC) exhibits similar biophysical, pharmacological, and molecular properties as I(CRAC) in nonexcitable cells and its rate of activation during repetitive depolarization is strongly regulated by temperature and RyR1 activity.


Calcium/metabolism , Ion Channel Gating , Muscle Fibers, Skeletal/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Temperature , Animals , Calcium Channels/metabolism , Membrane Glycoproteins/metabolism , Mice , ORAI1 Protein , Stromal Interaction Molecule 1 , Time Factors
14.
Nat Med ; 18(2): 244-51, 2012 Jan 08.
Article En | MEDLINE | ID: mdl-22231556

Mice with a knock-in mutation (Y524S) in the type I ryanodine receptor (Ryr1), a mutation analogous to the Y522S mutation that is associated with malignant hyperthermia in humans, die when exposed to short periods of temperature elevation (≥37 °C). We show here that treatment with 5-aminoimidazole-4-carboxamide ribonucleoside (AICAR) prevents this heat-induced sudden death in this mouse model. The protection by AICAR is independent of AMP-activated protein kinase (AMPK) activation and results from a newly identified action of the compound on mutant Ryr1 to reduce Ca(2+) leak from the sarcoplasmic reticulum to the sarcoplasm. AICAR thus prevents Ca(2+)-dependent increases in the amount of both reactive oxygen species (ROS) and reactive nitrogen species (RNS) that act to further increase resting Ca(2+) concentrations. If unchecked, the temperature-driven increases in resting Ca(2+) concentrations and the amounts of ROS and RNS create an amplifying cycle that ultimately triggers sustained muscle contractions, rhabdomyolysis and death. Although antioxidants are effective in reducing this cycle in vitro, only AICAR prevents heat-induced death in vivo. Our findings suggest that AICAR is probably effective in prophylactic treatment of humans with enhanced susceptibility to exercise- and/or heat-induced sudden death associated with RYR1 mutations.


AMP-Activated Protein Kinases/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Heat Stress Disorders/prevention & control , Hot Temperature/adverse effects , Ribonucleotides/pharmacology , Ryanodine Receptor Calcium Release Channel/genetics , AMP-Activated Protein Kinases/physiology , Adenosine Triphosphate/metabolism , Aminoimidazole Carboxamide/pharmacology , Animals , Calcium/metabolism , Death, Sudden/prevention & control , Enzyme Activation , Heat Stress Disorders/genetics , Male , Mice , Mice, Mutant Strains , Mice, Transgenic , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Ryanodine Receptor Calcium Release Channel/drug effects , Ryanodine Receptor Calcium Release Channel/physiology , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism
15.
Hum Mol Genet ; 21(6): 1312-24, 2012 Mar 15.
Article En | MEDLINE | ID: mdl-22140091

Myotonic dystrophy type 1 and type 2 (DM1 and DM2) are genetic diseases in which mutant transcripts containing expanded CUG or CCUG repeats cause cellular dysfunction by altering the processing or metabolism of specific mRNAs and miRNAs. The toxic effects of mutant RNA are mediated partly through effects on proteins that regulate alternative splicing. Here we show that alternative splicing of exon 29 (E29) of Ca(V)1.1, a calcium channel that controls skeletal muscle excitation-contraction coupling, is markedly repressed in DM1 and DM2. The extent of E29 skipping correlated with severity of weakness in tibialis anterior muscle of DM1 patients. Two splicing factors previously implicated in DM1, MBNL1 and CUGBP1, participated in the regulation of E29 splicing. In muscle fibers of wild-type mice, the Ca(V)1.1 channel conductance and voltage sensitivity were increased by splice-shifting oligonucleotides that induce E29 skipping. In contrast to human DM1, expression of CUG-expanded RNA caused only a modest increase in E29 skipping in mice. However, forced skipping of E29 in these mice, to levels approaching those observed in human DM1, aggravated the muscle pathology as evidenced by increased central nucleation. Together, these results indicate that DM-associated splicing defects alter Ca(V)1.1 function, with potential for exacerbation of myopathy.


Alternative Splicing , Calcium Channels, L-Type/physiology , Calcium/metabolism , Ion Channel Gating/physiology , Muscle Weakness/etiology , Myotonic Disorders/physiopathology , Myotonic Dystrophy/physiopathology , Animals , CELF1 Protein , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Exons/genetics , Gene Expression Regulation , Humans , Immunoblotting , Mice , Mice, Transgenic , Morpholinos/pharmacology , Muscle Weakness/metabolism , Muscle Weakness/pathology , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Myoblasts/cytology , Myoblasts/metabolism , Patch-Clamp Techniques , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction
16.
J Pharmacol Exp Ther ; 340(2): 463-72, 2012 Feb.
Article En | MEDLINE | ID: mdl-22088954

T-type calcium channels (Ca(V)3) play an important role in many physiological and pathological processes, including cancerogenesis. Ca(V)3 channel blockers have been proposed as potential cancer treatments. Roscovitine, a trisubstituted purine, is a cyclin-dependent kinase (CDK) inhibitor that is currently undergoing phase II clinical trials as an anticancer drug and has been shown to affect calcium and potassium channel activity. Here, we investigate the effect of roscovitine on Ca(V)3.1 channels. Ca(V)3.1 channels were transiently expressed in human embryonic kidney 293 cells, and currents were recorded by using the whole-cell patch-clamp technique. Roscovitine blocks Ca(V)3.1 channels with higher affinity for depolarized cells (EC50 of 10 µM), which is associated with a negative shift in the voltage dependence of closed-state inactivation. Enhanced inactivation is mediated by roscovitine-induced acceleration of closed-state inactivation and slowed recovery from inactivation. Small effects of roscovitine were also observed on T-channel deactivation and open-state inactivation, but neither could explain the inhibitory effect. Roscovitine inhibits Ca(V)3.1 channels within the therapeutic range (10-50 µM) in part by stabilizing the closed-inactivated state. The ability of roscovitine to block multiple mediators of proliferation, including CDKs and Ca(V)3.1 channels, may facilitate its anticancer properties.


Calcium Channels, T-Type/drug effects , Ion Channel Gating/drug effects , Purines/pharmacology , Action Potentials/physiology , Animals , Antineoplastic Agents/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/physiology , Dose-Response Relationship, Drug , Electric Stimulation , Electrophysiological Phenomena/drug effects , Electrophysiological Phenomena/physiology , HEK293 Cells , Humans , Ion Channel Gating/physiology , Kinetics , Membrane Potentials/physiology , Rats , Roscovitine , Transfection
17.
J Neurophysiol ; 107(7): 1942-51, 2012 Apr.
Article En | MEDLINE | ID: mdl-22205645

Ca(V)2.2 (N-type) and Ca(V)1.2 (L-type) calcium channels gate differently in response to membrane depolarization, which is critical to the unique physiological functions mediated by these channels. We wondered if the source for these differences could be identified. As a first step, we examined the effect of domain exchange between N-type and L-type channels on activation-deactivation kinetics, which were significantly different between these channels. Kinetic analysis of chimeric channels revealed N-channel-like deactivation for all chimeric channels containing N-channel domain III, while activation appeared to be a more distributed function across domains. This led us to hypothesize that domain III was an important regulator of N-channel closing. This idea was further examined with R-roscovitine, which is a trisubstituted purine that slows N-channel deactivation by exclusively binding to activated N-channels. L-channels lack this response to roscovitine, which allowed us to use N-L chimeras to test the role of domain III in roscovitine modulation of N-channel deactivation. In support of our hypothesis, all chimeric channels containing the N-channel domain III responded to roscovitine with slowed deactivation, while those chimeric channels with L-channel domain III did not. Thus a combination of kinetic and pharmacological evidence supports the hypothesis that domain III is an important regulator of N-channel closing. Our results support specialization of gating functions among calcium channel domains.


Biophysical Phenomena/physiology , Calcium Channels, N-Type/chemistry , Calcium Channels, N-Type/physiology , Ion Channel Gating/physiology , Animals , Biophysical Phenomena/drug effects , Biophysical Phenomena/genetics , Calcium Channels, N-Type/genetics , Electric Stimulation , HEK293 Cells , Humans , Ion Channel Gating/drug effects , Ion Channel Gating/genetics , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mutant Chimeric Proteins/genetics , Patch-Clamp Techniques , Protein Kinase Inhibitors/pharmacology , Protein Structure, Tertiary , Protein Subunits/genetics , Protein Subunits/metabolism , Purines/pharmacology , Rabbits , Roscovitine , Transfection
18.
J Clin Pharmacol ; 51(5): 719-30, 2011 May.
Article En | MEDLINE | ID: mdl-20547772

Propofol is commonly used to induce anesthesia but has been associated with some negative cardiovascular side effects, including negative inotropy, hypotension, and bradycardia. This study investigated the effect of propofol on L-type calcium current in acutely isolated human atrial myocytes to better understand the mechanism of these side effects. After informed consent was obtained, the atrial appendage was obtained from patients undergoing open-heart surgery who required cardiopulmonary bypass. Atrial myocytes were isolated using enzymatic digestion, and L-type calcium currents were recorded using the whole-cell patch clamp technique. Propofol enhanced the magnitude and speed of voltage-dependent inactivation of L-current. As a result, the propofol-induced inhibition was increased by protocols that increased inactivation such as longer voltage step duration, holding potential depolarization, and increased pulsing frequency. The preferential enhancement of L-channel inactivation by propofol can explain the associated cardiovascular side effects. The depolarized resting potential of arterial smooth muscle may render the L-channels in these cells particularly sensitive to propofol-induced inhibition, which could explain the hypotension observed in some patients. The enhancement of both inactivation kinetics and steady-state inactivation by propofol can also explain the negative inotropic effect. However, the enhanced voltage-dependent inactivation and use dependence could have beneficial effects for patients prone to certain arrhythmias and tachycardia.


Anesthetics, Intravenous/pharmacology , Atrial Appendage/drug effects , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/drug effects , Myocytes, Cardiac/drug effects , Propofol/pharmacology , Anesthetics, Intravenous/adverse effects , Atrial Appendage/metabolism , Bradycardia/chemically induced , Calcium Channel Blockers/adverse effects , Calcium Channels, L-Type/metabolism , Electric Stimulation , Humans , Hypotension/chemically induced , Kinetics , Membrane Potentials , Myocytes, Cardiac/metabolism , Patch-Clamp Techniques , Propofol/adverse effects
19.
Biochim Biophys Acta ; 1798(9): 1821-8, 2010 Sep.
Article En | MEDLINE | ID: mdl-20471360

N-type calcium channels play an important role in synaptic transmission and a drug that blocks these channels has become an important tool in controlling chronic pain. The development of new N-channel-targeted drugs is dependent on a better understanding of the gating of these channels and how that gating can be modulated. We have previously concluded that omega-conotoxin GVIA (GVIA) is a gating modifier that acts by destabilizing the N-channel open state. However, this conclusion was largely based on our modeling results and requires experimental support. Roscovitine, a tri-substituted purine, has been shown to stabilize the N-channel open state to slow gating charge relaxation, which provides a direct test of our hypothesis for GVIA-induced gating modification. We found that roscovitine could modulate gating current in the presence of GVIA, which shows that roscovitine can still affect the gating of the GVIA-bound N-channel. However, the magnitude of the roscovitine-induced slowing of Off-gating current was significantly reduced. In addition to confirming our hypothesis, our evidence supports an additional effect of GVIA to alter gating transitions between N-channel closed states. By strongly limiting access to the N-channel open state, GVIA analogs that selectively induce this modulation could provide the basis for the next generation drugs that treat chronic pain.


Calcium Channel Blockers/pharmacology , Calcium Channels, N-Type/drug effects , Ion Channel Gating/drug effects , omega-Conotoxin GVIA/pharmacology , Calcium Channels, N-Type/physiology , Cells, Cultured , Humans , Purines/pharmacology , Roscovitine
20.
Biophys J ; 98(7): 1149-59, 2010 Apr 07.
Article En | MEDLINE | ID: mdl-20371314

Ca(2+) entry through L-type calcium channels (Ca(V)1.2) is critical in shaping the cardiac action potential and initiating cardiac contraction. Modulation of Ca(V)1.2 channel gating directly affects myocyte excitability and cardiac function. We have found that phospholemman (PLM), a member of the FXYD family and regulator of cardiac ion transport, coimmunoprecipitates with Ca(V)1.2 channels from guinea pig myocytes, which suggests PLM is an endogenous modulator. Cotransfection of PLM in HEK293 cells slowed Ca(V)1.2 current activation at voltages near the threshold for activation, slowed deactivation after long and strong depolarizing steps, enhanced the rate and magnitude of voltage-dependent inactivation (VDI), and slowed recovery from inactivation. However, Ca(2+)-dependent inactivation was not affected. Consistent with slower channel closing, PLM significantly increased Ca(2+) influx via Ca(V)1.2 channels during the repolarization phase of a human cardiac action potential waveform. Our results support PLM as an endogenous regulator of Ca(V)1.2 channel gating. The enhanced VDI induced by PLM may help protect the heart under conditions such as ischemia or tachycardia where the channels are depolarized for prolonged periods of time and could induce Ca(2+) overload. The time and voltage-dependent slowed deactivation could represent a gating shift that helps maintain Ca(2+) influx during the cardiac action potential waveform plateau phase.


Calcium Channels, L-Type/metabolism , Heart/physiology , Membrane Proteins/metabolism , Phosphoproteins/metabolism , Action Potentials , Animals , Calcium/metabolism , Guinea Pigs , Heart Ventricles/pathology , Humans , Ions , Ischemia/pathology , Models, Biological , Muscle Cells/metabolism , Protein Structure, Tertiary , Tachycardia/pathology
...