Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Neuropharmacology ; 171: 108087, 2020 07.
Article in English | MEDLINE | ID: mdl-32272140

ABSTRACT

Cyclic nucleotide-gated (CNG) channels, which are directly activated by cAMP and cGMP, have long been known to play a key role in retinal and olfactory signal transduction. Emerging evidence indicates that CNG channels are also involved in signaling pathways important for pain processing. Here, we found that the expression of the channel subunits CNGA2, CNGA3, CNGA4 and CNGB1 in dorsal root ganglia, and of CNGA2 in the spinal cord, is transiently altered after peripheral nerve injury in mice. Specifically, we show using in situ hybridization and quantitative real-time RT-PCR that CNG channels containing the CNGB1b subunit are localized to populations of sensory neurons and predominantly excitatory interneurons in the spinal dorsal horn. In CNGB1 knockout (CNGB1-/-) mice, neuropathic pain behavior is considerably attenuated whereas inflammatory pain behavior is normal. Finally, we provide evidence to support CNGB1 as a downstream mediator of cAMP signaling in pain pathways. Altogether, our data suggest that CNGB1-positive CNG channels specifically contribute to neuropathic pain processing after peripheral nerve injury.


Subject(s)
Cyclic AMP , Cyclic Nucleotide-Gated Cation Channels/genetics , Nerve Tissue Proteins/genetics , Neuralgia/psychology , Pain/chemically induced , Pain/psychology , Animals , Cyclic Nucleotide-Gated Cation Channels/biosynthesis , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Inflammation/chemically induced , Inflammation/pathology , Injections, Spinal , Mice, Inbred C57BL , Mice, Knockout , Neuralgia/pathology , Pain/pathology , Postural Balance/drug effects , Signal Transduction/drug effects , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology
2.
Invest Ophthalmol Vis Sci ; 57(3): 787-97, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26934134

ABSTRACT

PURPOSE: Rod photoreceptor outer segment (OS) morphogenesis, structural integrity, and proper signal transduction rely on critical proteins found in the different OS membrane domains (e.g., plasma, disc, and disc rim membrane). Among these key elements are retinal degeneration slow (RDS, also known as peripherin-2), rhodopsin, and the beta subunit of the cyclic nucleotide gated channel (CNGB1a), which have been found to interact in a complex. The purpose of this study was to evaluate the potential interplay between these three proteins by examining retinal disease phenotypes in animal models expressing varying amounts of CNGB1a, rhodopsin, and RDS. METHODS: Outer segment trafficking, retinal function, and photoreceptor structure were evaluated using knockout mouse lines. RESULTS: Eliminating Cngb1 and reducing RDS leads to additive defects in RDS expression levels and rod electroretinogram (ERG) function, (e.g., Cngb1-/-/rds+/- versus rds+/- or Cngb1-/-) but not to additive defects in rod ultrastructure. These additive effects also manifested in cone function: Photopic ERG responses were significantly lower in the Cngb1-/-/rds+/- versus rds+/- or Cngb1-/-, suggesting that eliminating Cngb1 can accelerate the cone degeneration that usually presents later in the rds+/-. This was not the case with rhodopsin; reducing rhodopsin levels in concert with eliminating CNGB1a did not lead to phenotypes more severe than those observed in the Cngb1 knockout alone. CONCLUSIONS: These data support a role for RDS as the core component of a multiprotein plasma membrane-rim-disc complex that has both a structural role in photoreceptor OS formation and maintenance and a functional role in orienting proteins for optimal signal transduction.


Subject(s)
Cyclic Nucleotide-Gated Cation Channels/genetics , Gene Expression Regulation , Nerve Tissue Proteins/genetics , Peripherins/genetics , RNA/genetics , Retinal Cone Photoreceptor Cells/metabolism , Retinal Degeneration/genetics , Rhodopsin/genetics , Animals , Cyclic Nucleotide-Gated Cation Channels/biosynthesis , Electroretinography , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Nerve Tissue Proteins/biosynthesis , Peripherins/biosynthesis , Retinal Cone Photoreceptor Cells/ultrastructure , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Rhodopsin/biosynthesis
3.
PLoS One ; 9(2): e88768, 2014.
Article in English | MEDLINE | ID: mdl-24586388

ABSTRACT

Cone cyclic nucleotide-gated channels are tetramers formed by CNGA3 and CNGB3 subunits; CNGA3 subunits function as homotetrameric channels but CNGB3 exhibits channel function only when co-expressed with CNGA3. An aspartatic acid (Asp) to asparagine (Asn) missense mutation at position 262 in the canine CNGB3 (D262N) subunit results in loss of cone function (daylight blindness), suggesting an important role for this aspartic acid residue in channel biogenesis and/or function. Asp 262 is located in a conserved region of the second transmembrane segment containing three Asp residues designated the Tri-Asp motif. This motif is conserved in all CNG channels. Here we examine mutations in canine CNGA3 homomeric channels using a combination of experimental and computational approaches. Mutations of these conserved Asp residues result in the absence of nucleotide-activated currents in heterologous expression. A fluorescent tag on CNGA3 shows mislocalization of mutant channels. Co-expressing CNGB3 Tri-Asp mutants with wild type CNGA3 results in some functional channels, however, their electrophysiological characterization matches the properties of homomeric CNGA3 channels. This failure to record heteromeric currents suggests that Asp/Asn mutations affect heteromeric subunit assembly. A homology model of S1-S6 of the CNGA3 channel was generated and relaxed in a membrane using molecular dynamics simulations. The model predicts that the Tri-Asp motif is involved in non-specific salt bridge pairings with positive residues of S3/S4. We propose that the D262N mutation in dogs with CNGB3-day blindness results in the loss of these inter-helical interactions altering the electrostatic equilibrium within in the S1-S4 bundle. Because residues analogous to Tri-Asp in the voltage-gated Shaker potassium channel family were implicated in monomer folding, we hypothesize that destabilizing these electrostatic interactions impairs the monomer folding state in D262N mutant CNG channels during biogenesis.


Subject(s)
Amino Acid Motifs/genetics , Cyclic Nucleotide-Gated Cation Channels/genetics , Dog Diseases/genetics , Dog Diseases/pathology , Mutation, Missense/genetics , Retinal Cone Photoreceptor Cells/pathology , Retinal Degeneration/veterinary , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Computational Biology/methods , Cyclic Nucleotide-Gated Cation Channels/biosynthesis , DNA Primers/genetics , Dogs , Fluorescence , Immunohistochemistry , Models, Genetic , Molecular Dynamics Simulation , Molecular Sequence Data , Retinal Degeneration/genetics , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology
4.
Circulation ; 127(20): 2009-20, 2013 May 21.
Article in English | MEDLINE | ID: mdl-23609807

ABSTRACT

BACKGROUND: Pregnancy is associated with a faster heart rate (HR), which is a risk factor for arrhythmias. However, the underlying mechanisms for this increased HR are poorly understood. Therefore, this study was performed to gain mechanistic insight into the pregnancy-induced increase in HR. METHODS AND RESULTS: Using surface ECG we observed that pregnant (P) mice have faster HR (531±14 beats per minute [bpm]) compared with nonpregnant (NP) mice (470±27 bpm; P<0.03). Results obtained with Langendorff-perfused hearts showed that this difference persisted in the absence of autonomic nervous innervation (NP, 327±16 bpm; P, 385±18 bpm; P<0.02). Spontaneous action potentials of sinoatrial node cells from pregnant mice exhibited higher automaticity (NP, 292±13 bpm; P, 330±12 bpm; P=0.047) and steeper diastolic depolarization (NP, 0.20±0.03 V/s; P, 0.40±0.06 V/s; P=0.004). Pregnancy increased the density of the hyperpolarization-activated current (If) (at -90mV: NP, -15.2±1.0 pA/pF; P, -28.6±2.9 pA/pF; P=0.0002) in sinoatrial node cells. Voltage dependence of the If activation curve and the intracellular cAMP levels were unchanged in sinoatrial node cells of pregnant mice. However, there was a significant increase in HCN2 channel protein expression with no change in HCN4 expression. Maximal depolarizing shift of the If activation curve induced by isoproterenol was attenuated in pregnancy. This reduced response to isoproterenol may be attributable to the lower cAMP sensitivity of HCN2 isoform compared with that of HCN4. CONCLUSIONS: This study shows that an increase in If current density contributes to the acceleration of sinoatrial node automaticity and explains, in part, the higher HR observed in pregnancy.


Subject(s)
Heart Conduction System/physiology , Heart Rate/physiology , Ion Channels/biosynthesis , Pregnancy/physiology , Sinoatrial Node/physiology , Up-Regulation/physiology , Animals , Cyclic Nucleotide-Gated Cation Channels/biosynthesis , Female , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Mice , Potassium Channels
5.
Neuron ; 75(3): 503-16, 2012 Aug 09.
Article in English | MEDLINE | ID: mdl-22884333

ABSTRACT

The hippocampus is an integral brain region for affective disorders. TRIP8b knockout mice lacking functional HCN channels as well as both HCN1 and HCN2 knockout mice have been shown to display antidepressant-like behaviors. The mechanisms or brain regions involved in these alterations in behavior, however, are not clear. We developed a lentiviral shRNA system to examine whether knockdown of HCN1 protein in the dorsal hippocampal CA1 region is sufficient to produce antidepressant-like effects. We found that knockdown of HCN1 channels increased cellular excitability and resulted in physiological changes consistent with a reduction of I(h). Rats infused with lentiviral shRNA-HCN1 in the dorsal hippocampal CA1 region displayed antidepressant- and anxiolytic-like behaviors associated with widespread enhancement of hippocampal activity and upregulation of BDNF-mTOR signaling pathways. Our results suggest that HCN1 protein could be a potential target for treatment of anxiety and depression disorders.


Subject(s)
Anxiety/genetics , Cyclic Nucleotide-Gated Cation Channels/genetics , Depression/genetics , Hippocampus/metabolism , Potassium Channels/genetics , Animals , Anxiety/metabolism , Behavior, Animal , Blotting, Western , Cyclic Nucleotide-Gated Cation Channels/biosynthesis , Depression/metabolism , Gene Knockdown Techniques , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Immunohistochemistry , Male , Patch-Clamp Techniques , Potassium Channels/biosynthesis , RNA, Small Interfering , Rats , Rats, Sprague-Dawley
6.
Urology ; 80(1): 224.e13-8, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22748890

ABSTRACT

OBJECTIVE: To investigate the expression of hyperpolarization-activated cyclic nucleotide-gated (HCN) protein in the interstitial cells of Cajal (ICCs) of the human bladder and to determine the relative expression of the HCN subtypes. METHODS: A total of 30 bladder specimens were obtained, and each bladder sample was divided into 2 parts. Part I, which included mucosa, submucosa, and muscle, was used for immunofluorescence study. Part II, in which the mucosa and submucosa were removed and fresh bladder muscle tissue was kept, was used for Western blotting. First, the immunoreactivity of HCN1, HCN2, HCN3, and HCN4 isoforms was detected in the ICCs of lamina propria using immunofluorescence. Second, the protein expression of HCN1, HCN2, HCN3, and HCN4 isoforms in ICCs of bladder detrusor muscle was analyzed using Western blotting. RESULTS: Immunofluorescence showed that there were many vimentin- and HCN4-positive ICCs in the lamina propria region and detrusor. Also, only a few c-kit and HCN4-positive ICCs were detected in the lamina propria region and detrusor. Additionally, novel c-kit negative, but HCN4-positive mast cells were found in the lamina propria. Immunofluorescence and Western blotting revealed that HCN1, HCN2, HCN3, and HCN4 channels were all present in the ICCs of the human bladder; however, HCN4 channel expression in the ICCs was greater than in HCN1, HCN2, or HCN3. CONCLUSION: HCN1, HCN2, HCN3, and HCN4 channels are identified in ICCs of human bladder tissue, and the expression of the HCN4 channel exceeded that of the other subtypes.


Subject(s)
Cyclic Nucleotide-Gated Cation Channels/biosynthesis , Interstitial Cells of Cajal/metabolism , Urinary Bladder/cytology , Humans
7.
J Biol Chem ; 287(21): 17656-17661, 2012 May 18.
Article in English | MEDLINE | ID: mdl-22511771

ABSTRACT

The dorsal and ventral regions of the hippocampus perform different functions. Whether the integrative properties of hippocampal cells reflect this heterogeneity is unknown. We focused on dendrites where most synaptic input integration takes place. We report enhanced backpropagation and theta resonance and decreased summation of synaptic inputs in ventral versus dorsal CA1 pyramidal cell distal dendrites. Transcriptional Kv4.2 down-regulation and post-transcriptional hyperpolarization-activated cyclic AMP-gated channel (HCN1/2) up-regulation may underlie these differences, respectively. Our results reveal differential dendritic integrative properties along the dorso-ventral axis, reflecting diverse computational needs.


Subject(s)
Cyclic Nucleotide-Gated Cation Channels/biosynthesis , Dendrites/metabolism , Down-Regulation/physiology , Ion Channels/biosynthesis , Nerve Tissue Proteins/biosynthesis , Potassium Channels/biosynthesis , Pyramidal Cells/metabolism , Shal Potassium Channels/biosynthesis , Up-Regulation/physiology , Animals , Dendrites/genetics , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Organ Specificity , Pyramidal Cells/cytology , Rats , Transcription, Genetic/physiology
8.
Biochem Biophys Res Commun ; 420(1): 156-60, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22405820

ABSTRACT

The hyperpolarization-activated cation current I(h) is an important regulator of neuronal excitability and may contribute to the properties of the dentate gyrus granule (DGG) cells, which constitute the input site of the canonical hippocampal circuit. Here, we investigated changes in I(h) in DGG cells in human temporal lobe epilepsy (TLE) and the rat pilocarpine model of TLE using the patch-clamp technique. Messenger-RNA (mRNA) expression of I(h)-conducting HCN1, 2 and 4 isoforms was determined using semi-quantitative in-situ hybridization. I(h) density was ∼1.8-fold greater in DGG cells of TLE patients with Ammon's horn sclerosis (AHS) as compared to patients without AHS. The magnitude of somatodendritic I(h) was enhanced also in DGG cells in epileptic rats, most robustly during the latent phase after status epilepticus and prior to the occurrence of spontaneous epileptic seizures. During the chronic phase, I(h) was increased ∼1.7-fold. This increase of I(h) was paralleled by an increase in HCN1 and HCN4 mRNA expression, whereas HCN2 expression was unchanged. Our data demonstrate an epilepsy-associated upregulation of I(h) likely due to increased HCN1 and HCN4 expression, which indicate plasticity of I(h) during epileptogenesis and which may contribute to a compensatory decrease in neuronal excitability of DGG cells.


Subject(s)
Cyclic Nucleotide-Gated Cation Channels/biosynthesis , Dentate Gyrus/physiopathology , Epilepsy, Temporal Lobe/physiopathology , Potassium Channels/biosynthesis , Animals , Cells, Cultured , Cyclic Nucleotide-Gated Cation Channels/chemical synthesis , Dentate Gyrus/metabolism , Disease Models, Animal , Epilepsy, Temporal Lobe/metabolism , Epilepsy, Temporal Lobe/therapy , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Neurons/metabolism , Neurons/physiology , Patch-Clamp Techniques , Pilocarpine/pharmacology , Potassium Channels/chemical synthesis , Rats , Up-Regulation
9.
Mol Endocrinol ; 26(1): 153-64, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22135067

ABSTRACT

Pituitary cells fire action potentials independently of external stimuli, and such spontaneous electrical activity is modulated by a large variety of hypothalamic and intrapituitary agonists. Here, we focused on the potential role of hyperpolarization-activated and cyclic nucleotide-gated (HCN) channels in electrical activity of cultured rat anterior pituitary cells. Quantitative RT-PCR analysis showed higher level of expression of mRNA transcripts for HCN2 and HCN3 subunits and lower expression of HCN1 and HCN4 subunits in these cells. Western immunoblot analysis of lysates from normal and GH(3) immortalized pituitary cells showed bands with appropriate molecular weights for HCN2, HCN3, and HCN4. Electrophysiological experiments showed the presence of a slowly developing hyperpolarization-activated inward current, which was blocked by Cs(+) and ZD7288, in gonadotrophs, thyrotrophs, somatotrophs, and a fraction of lactotrophs, as well as in other unidentified pituitary cell types. Stimulation of adenylyl cyclase and addition of 8-Br-cAMP enhanced this current and depolarized the cell membrane, whereas 8-Br-cGMP did not alter the current and hyperpolarized the cell membrane. Both inhibition of basal adenylyl cyclase activity and stimulation of phospholipase C signaling pathway inhibited this current. Inhibition of HCN channels affected the frequency of firing but did not abolish spontaneous electrical activity. These experiments indicate that cAMP and cGMP have opposite effects on the excitability of endocrine pituitary cells, that basal cAMP production in cultured cells is sufficient to integrate the majority of HCN channels in electrical activity, and that depletion of phosphatidylinositol 4,5-bisphosphate caused by activation of phospholipase C silences them.


Subject(s)
Cyclic Nucleotide-Gated Cation Channels/metabolism , Endocrine Cells/metabolism , Pituitary Gland, Anterior/metabolism , Potassium Channels/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Action Potentials/drug effects , Adenylyl Cyclases/biosynthesis , Animals , Cell Membrane/metabolism , Cells, Cultured , Cesium/pharmacology , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Cyclic Nucleotide-Gated Cation Channels/biosynthesis , Female , Gonadotrophs/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Lactotrophs/metabolism , Membrane Potentials/drug effects , Phosphatidylinositol 4,5-Diphosphate/deficiency , Pituitary Gland, Anterior/cytology , Potassium Channels/biosynthesis , Pyrimidines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Somatotrophs/metabolism , Thyrotrophs/metabolism , Type C Phospholipases/biosynthesis , Type C Phospholipases/metabolism
10.
J Neurochem ; 118(6): 988-98, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21749376

ABSTRACT

TPR-containing Rab8b-interacting protein (TRIP8b) is a brain-specific hydrophilic cytosolic protein that contains tetratricopeptide repeats (TPRs). Previous studies revealed interaction of this protein via its TPR-containing domain with Rab8b small GTPase, hyperpolarization-activated cyclic nucleotide-regulated channel (HCN) channels and G protein-coupled receptor calcium-independent receptor of α-latrotoxin. We identified clathrin as a major component of eluates from the TRIP8b affinity matrix. In the present study, by in vitro-binding analysis we demonstrate a direct interaction between clathrin and TRIP8b. The clathrin-binding site was localized in the N-terminal (non-TPR containing) part of the TRIP8b molecule that contains two short motifs involved in the clathrin binding. In transfected HEK293 cells, co-expression of HCN1 with TRIP8b resulted in translocation of the channels from the cell surface to large intracellular puncta where both TRIP8b and clathrin were concentrated. These puncta co-localized partially with an early endosome marker and strongly overlapped with lysosome staining reagent. When HCN1 was co-expressed with a clathrin-non-binding mutant of TRIP8b, clathrin did not translocate to HCN1 and TRIP8b-containing puncta, suggesting that TRIP8b interacts with HCN and clathrin independently. We found TRIP8b present in the fraction of clathrin-coated vesicles purified from brain tissues. Stripping the clathrin coat proteins from the vesicles with Tris alkaline buffer resulted in concomitant release of TRIP8b. Our data suggest complex regulatory functions of TRIP8b in neuronal endocytosis through independent interaction with membrane proteins and components of the clathrin coat.


Subject(s)
Clathrin/biosynthesis , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Animals , Blotting, Western , Cell Line , Clathrin/genetics , Clathrin/isolation & purification , Cyclic Nucleotide-Gated Cation Channels/biosynthesis , Cyclic Nucleotide-Gated Cation Channels/genetics , DNA/biosynthesis , DNA/genetics , Electrophoresis, Polyacrylamide Gel , Endocytosis , Escherichia coli/metabolism , Exons/genetics , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Immunohistochemistry , Mass Spectrometry , Membrane Proteins/isolation & purification , Plasmids/genetics , Point Mutation , Potassium Channels/biosynthesis , Potassium Channels/genetics , Protein Binding , Rats , Subcellular Fractions/metabolism
11.
J Neurophysiol ; 106(4): 2045-56, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21753027

ABSTRACT

Hyperpolarization-activated inward currents (I(h)) contribute to neuronal excitability in sensory neurons. Four subtypes of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels generate I(h), with different activation kinetics and cAMP sensitivities. The aim of the present study was to examine the postnatal development of I(h) and HCN channel subunits in trigeminal ganglion (TG) neurons. I(h) was investigated in acutely dissociated TG neurons from rats aged between postnatal day (P)1 and P35 with whole cell patch-clamp electrophysiology. In voltage-clamp studies, I(h) was activated by a series of hyperpolarizing voltage steps from -40 mV to -120 mV in -10-mV increments. Tail currents from a common voltage step (-100 mV) were used to determine I(h) voltage dependence. I(h) activation was faster in older rats and occurred at more depolarized potentials; the half-maximal activation voltage (V(1/2)) changed from -89.4 mV (P1) to -81.6 mV (P35). In current-clamp studies, blocking I(h) with ZD7288 caused membrane hyperpolarization and increases in action potential half-duration at all postnatal ages examined. ZD7288 also reduced the action potential firing frequency in multiple-firing neurons. Western blot analysis of the TG detected immunoreactive bands corresponding to all HCN subtypes. HCN1 and HCN2 band density increased with postnatal age, whereas the low-intensity HCN3 and moderate-intensity HCN4 bands were not changed. This study suggests that functional I(h) are activated in rat trigeminal sensory neurons from P1 during postnatal development, have an increasing role with age, and modify neuronal excitability.


Subject(s)
Cyclic Nucleotide-Gated Cation Channels/biosynthesis , Gene Expression Regulation, Developmental , Potassium Channels/biosynthesis , Sensory Receptor Cells/physiology , Trigeminal Ganglion/growth & development , Amino Acid Sequence , Animals , Blotting, Western , Cyclic Nucleotide-Gated Cation Channels/genetics , Cyclic Nucleotide-Gated Cation Channels/physiology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Ion Channels , Molecular Sequence Data , Nociception/physiology , Patch-Clamp Techniques , Potassium Channels/genetics , Potassium Channels/physiology , Protein Subunits , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley , Trigeminal Ganglion/cytology , Trigeminal Ganglion/metabolism
12.
Cardiovasc Pathol ; 20(2): 110-3, 2011.
Article in English | MEDLINE | ID: mdl-20207172

ABSTRACT

OBJECTIVE: Hypertrophic cardiomyopathy (HCM) is a disease of the myocardium with uncertain etiology and often leads to sudden death as the result of arrhythmia. Pacemaker hyperpolarization-activated current I(f) was altered in hypertrophic hearts and was probably responsible for arrhythmia. I(f) channels are compose\d of four hyperpolarization-activated cyclic nucleotide-gated cation subunits (HCN1-4). A previous study found significantly high levels of HCN2 and HCN4 mRNA in hypertrophic hearts compared to control hearts in septum and left ventricles in rats. No studies, however, have investigated the HCN gene expression in the myocardium from human HCM heart. METHODS: The left ventricular tissue from four patients who died of HCM and six healthy patients who died of motor vehicle accidents was included in this study. The fluorescent quantitative reverse transcription-polymerase chain reaction (RT-PCR) assay was used to detect HCN4 mRNA. The expression of HCN4 mRNA of the two groups was detected on the assay. RESULTS: In the HCM hearts, disorganization of the hypertrophic myofibers and interstitial fibrosis were observed in all four patients, although absent in healthy control hearts. By quantitative polymerase chain reaction, the mean copy number of HCN4 mRNA was 2.2×10(7) (range, 6.8×10(6) to 4.55×10(7)) in HCM hearts and 8.17×10(3) (range, 8.76×10(1) to 3.5×10(4)) in control hearts (P=.0318). CONCLUSION: Higher HCN4 mRNA levels in the HCM hearts suggest that up-regulation of HCN4 gene expression might be responsible for ventricular arrhythmia that leads to sudden death.


Subject(s)
Cardiomyopathy, Hypertrophic/metabolism , Cyclic Nucleotide-Gated Cation Channels/biosynthesis , Muscle Proteins/biosynthesis , Adult , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/pathology , Cyclic Nucleotide-Gated Cation Channels/genetics , Female , Gene Expression , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Male , Muscle Proteins/genetics , Myocardium/chemistry , Myocardium/metabolism , Potassium Channels , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
13.
Epilepsia ; 51 Suppl 3: 52-5, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20618401

ABSTRACT

Cortical dysplasia is often associated with intractable seizures. Studies in animal models have described changes in inhibitory and excitatory synaptic function that contribute to hyperexcitability. The role of changes in intrinsic excitability and abnormal dendritic properties has received less attention. Changes in hyperpolarization-activated nonselective cation (HCN) channels have been implicated in several models of epilepsy. Herein we review evidence for alterations in HCN channels and dendritic morphology in the rat freeze-lesion model of cortical dysplasia. Immunocytochemical HCN1 staining, typically seen in the apical dendrites of layer V pyramidal cells in normal cortex, was greatly reduced in the region adjacent to the freeze-induced microgyrus. Although staining was preserved in layer I, fewer dendrites were stained in upper cortical layers. Deeper cortical layers were virtually devoid of immunoreactivity. Examination of biocytin-labeled pyramidal cells revealed markedly altered dendritic trees in the lesioned animals. In addition, resting membrane properties were altered and a subpopulation of neurons with abnormal dendritic arbors was present. These changes are likely to interact with the previously reported synaptic changes in this model of cortical dysplasia. HCN channel alterations are a potentially important cellular mechanism underlying hyperexcitability in cortical dysplasia.


Subject(s)
Cyclic Nucleotide-Gated Cation Channels/physiology , Epilepsy/etiology , Malformations of Cortical Development/pathology , Potassium Channels/physiology , Pyramidal Cells/pathology , Animals , Animals, Newborn , Cyclic Nucleotide-Gated Cation Channels/biosynthesis , Disease Models, Animal , Epilepsy/pathology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Malformations of Cortical Development/complications , Potassium Channels/biosynthesis , Pyramidal Cells/physiopathology , Rats , Rats, Sprague-Dawley
14.
Neuroscience ; 165(1): 39-52, 2010 Jan 13.
Article in English | MEDLINE | ID: mdl-19815055

ABSTRACT

Vagal afferent neurons, serving as the primary afferent limb of the parasympathetic reflex, could be involved in diabetic autonomic neuropathy. Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are expressed in the vagal afferent neurons and play an important role in determining cell membrane excitation. In the present study, the protein expression and the electrophysiological characteristics of HCN channels were investigated in nodose ganglion (NG) afferent neurons (A-fiber and C-fiber neurons) from sham and streptozotocin (STZ)-induced diabetic rats. In the sham NG, HCN1, HCN3, and HCN4 were expressed in the A-fiber neurons; and HCN2, HCN3, and HCN4 were expressed in the C-fiber neurons. Compared to the sham NG neurons, diabetes induced the expression of HCN2 in the A-fiber neurons besides overexpression of HCN1 and HCN3; and enhanced the expression of HCN2 and HCN3 in C-fiber neurons. In addition, whole-cell patch-clamp data revealed diabetes also increased HCN currents in A-fiber and C-fiber neurons. However, we found that diabetes did not alter the total nodose afferent neuron number and the ratio of A-fiber/C-fiber neurons. These results indicate that diabetes induces the overexpression of HCN channels and the electrophysiological changes of HCN currents in the A- and C-fiber nodose neurons, which might contribute to the diabetes-induced alteration of cell excitability in the vagal afferent neurons.


Subject(s)
Cyclic Nucleotide-Gated Cation Channels/biosynthesis , Diabetes Mellitus, Experimental/metabolism , Neurons, Afferent/metabolism , Nodose Ganglion/metabolism , Animals , Cell Count , Cyclic AMP/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/physiopathology , Fluorescent Antibody Technique , Male , Nerve Fibers, Myelinated/metabolism , Nerve Fibers, Unmyelinated/metabolism , Neurons, Afferent/pathology , Neurons, Afferent/physiology , Nodose Ganglion/cytology , Patch-Clamp Techniques , Protein Isoforms/biosynthesis , Protein Subunits/biosynthesis , Rats , Rats, Sprague-Dawley
15.
Am J Physiol Renal Physiol ; 297(5): F1353-60, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19710238

ABSTRACT

We have addressed the distribution of cGMP-gated channels (CNG) in the rat urethra for the first time, as well as their putative role in mediating of the relaxation elicited by electrical field stimulation of nitrergic nerves. Functional studies have shown that specifically blocking CNG with L-cis-diltiazem leads to the rapid inhibition of urethral relaxation induced either by nitric oxide (NO) released by the nerves or by soluble guanylate cyclase activated with YC-1. By contrast, nerve-mediated noradrenergic contractions were only slowly and mildly reduced by L-cis-diltiazem. This effect was mimicked by lower concentrations of the D-diltiazem isomer, probably due to the nonspecific inhibition of voltage-dependent calcium channels. However, D-diltiazem did not affect relaxation responses. The expression of heteromeric retinal-like CNGA1 channels was demonstrated by conventional PCR on mRNA from the rat urethra. These channels were located in a subpopulation of intramuscular interstitial cells of Cajal (ICC) as well as in smooth muscle cells, although they were less abundant in the latter. CNG channels could not be visualized in any nervous structure within the urethral wall, in agreement with the emerging view that a subset of ICC serves as a target for NO. These channels could provide a suitable ionic mechanism to associate the changes in cytosolic calcium with the activation of the nitric NO-cGMP pathway and relaxation although the precise mechanisms involved remain to be elucidated.


Subject(s)
Cyclic Nucleotide-Gated Cation Channels/physiology , Nitric Oxide/physiology , Urethra/innervation , Urethra/metabolism , Animals , Calcium Channel Blockers/chemistry , Calcium Channel Blockers/pharmacology , Cyclic Nucleotide-Gated Cation Channels/antagonists & inhibitors , Cyclic Nucleotide-Gated Cation Channels/biosynthesis , Cyclic Nucleotide-Gated Cation Channels/genetics , Diltiazem/chemistry , Diltiazem/pharmacology , Enzyme Inhibitors/pharmacology , Female , Fluorescent Antibody Technique , Indazoles/pharmacology , Isometric Contraction/drug effects , Muscle Contraction/physiology , Muscle Relaxation/physiology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Stereoisomerism
16.
Pflugers Arch ; 458(6): 1061-8, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19629520

ABSTRACT

Thyroid hormone (TH) markedly modulates cardiovascular function and heart rate. The pacemaker current I(f) and encoding hyperpolarization-activated cation (HCN) genes have been identified as TH targets. To analyze the specific contribution and functional significance of thyroid receptor isoforms responsible for HCN gene transactivation, we generated transgenic neonatal rat cardiomyocytes with adenovirus-mediated overexpression of the thyroid receptors alpha1 (TR alpha 1) and beta1 (TR beta 1), and analyzed native I(f) current and expression levels of the underlying molecular components HCN2 and HCN4. Initial results revealed that spontaneous beating activity was higher in TR alpha 1- and lower in TR beta 1-expressing cardiomyocytes. This was associated with accelerated depolarization velocity and abbreviated action potential duration in cells overexpressing TR alpha 1, while TR beta 1 suppressed phase 4 depolarization and prolonged action potentials. Consistently, TR alpha 1-infected myocytes exhibited larger I(f) current densities along with increased HCN2 and HCN4 mRNA and protein levels. In contrast, HCN2 gene expression was not significantly affected by TR beta 1. TR beta 1 exclusively suppressed HCN4 transcription. T3 application led to significant effects only in controls and TR alpha 1-infected cardiomyocytes; whereas, no ligand-dependent actions were observed in TR beta 1-expressing neonatal cardiomyocytes. Our results demonstrate that TR alpha 1 and TR beta 1 divergently regulate cardiac pacing activity. TH-induced positive chronotropic effects are likely to be mediated by TR alpha 1 through enhanced expression of I(f) pacemaker current and its underlying genes.


Subject(s)
Cyclic Nucleotide-Gated Cation Channels/genetics , Heart/physiology , Ion Channels/genetics , Muscle Proteins/genetics , Potassium Channels/genetics , Thyroid Hormone Receptors alpha/physiology , Thyroid Hormone Receptors beta/physiology , Action Potentials/drug effects , Animals , Cyclic Nucleotide-Gated Cation Channels/biosynthesis , Electrophysiological Phenomena , Heart Rate/drug effects , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Ion Channels/biosynthesis , Muscle Proteins/biosynthesis , Myocytes, Cardiac/metabolism , Potassium Channels/biosynthesis , Rats
17.
J Neurosci ; 29(27): 8847-57, 2009 Jul 08.
Article in English | MEDLINE | ID: mdl-19587292

ABSTRACT

Hyperpolarization-activated cyclic nucleotide-gated cation (HCN) channels are the molecular substrate of the hyperpolarization-activated inward current (I(h)). Because the developmental profile of HCN channels in the thalamus is not well understood, we combined electrophysiological, molecular, immunohistochemical, EEG recordings in vivo, and computer modeling techniques to examine HCN gene expression and I(h) properties in rat thalamocortical relay (TC) neurons in the dorsal part of the lateral geniculate nucleus and the functional consequence of this maturation. Recordings of TC neurons revealed an approximate sixfold increase in I(h) density between postnatal day 3 (P3) and P106, which was accompanied by significantly altered current kinetics, cAMP sensitivity, and steady-state activation properties. Quantification on tissue levels revealed a significant developmental decrease in cAMP. Consequently the block of basal adenylyl cyclase activity was accompanied by a hyperpolarizing shift of the I(h) activation curve in young but not adult rats. Quantitative analyses of HCN channel isoforms revealed a steady increase of mRNA and protein expression levels of HCN1, HCN2, and HCN4 with reduced relative abundance of HCN4. Computer modeling in a simplified thalamic network indicated that the occurrence of rhythmic delta activity, which was present in the EEG at P12, differentially depended on I(h) conductance and modulation by cAMP at different developmental states. These data indicate that the developmental increase in I(h) density results from increased expression of three HCN channel isoforms and that isoform composition and intracellular cAMP levels interact in determining I(h) properties to enable progressive maturation of rhythmic slow-wave sleep activity patterns.


Subject(s)
Biological Clocks/physiology , Cerebral Cortex/metabolism , Cyclic Nucleotide-Gated Cation Channels/biosynthesis , Gene Expression Regulation, Developmental/physiology , Ion Channels/biosynthesis , Neurons/metabolism , Potassium Channels/biosynthesis , Thalamus/metabolism , Animals , Animals, Newborn , Cerebral Cortex/growth & development , Cyclic Nucleotide-Gated Cation Channels/genetics , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Ion Channels/genetics , Neural Pathways/growth & development , Neural Pathways/metabolism , Neurons/physiology , Potassium Channels/genetics , Protein Isoforms/biosynthesis , Rats , Rats, Sprague-Dawley , Thalamus/growth & development
18.
Basic Res Cardiol ; 104(6): 621-9, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19421833

ABSTRACT

Hyperpolarization-activated ion channels, encoded by four mammalian genes (HCN1-4), contribute in an important way to the cardiac pacemaker current I(f). Here, we describe the transcription profiles of the four HCN genes, the NRSF, KCNE2 and Kir2.1 genes from embryonic stage E9.5 dpc to postnatal day 120 in the mouse. Embryonic atrium and ventricle revealed abundant HCN4 transcription but other HCN transcripts were almost absent. Towards birth, HCN4 was downregulated in the atrium and almost vanished from the ventricle. After birth, however, HCN isotype transcription changed remarkably, showing increased levels of HCN1, HCN2 and HCN4 in the atrium and of HCN2 and HCN4 in the ventricle. HCN3 showed highest transcription at early embryonic stages and was hardly detectable thereafter. At postnatal day 10, HCN4 was highest in the sinoatrial node, being twofold higher than HCN1 and fivefold higher than HCN2. In the atrium, HCN4 was similar to HCN1 and sevenfold higher than HCN2. In the ventricle, in contrast, HCN2 was sixfold higher than HCN4, while HCN1 was absent. Subsequently all HCN isotype transcripts declined to lower adult levels, while ratios of HCN isotypes remained stable. In conclusion, substantial changes of HCN isotype transcription throughout cardiac development suggest that a regulated pattern of HCN isotypes is required to establish and ensure a stable heart rhythm. Furthermore, constantly low HCN transcription in adult myocardium may be required to prevent atrial and ventricular arrhythmogenesis.


Subject(s)
Heart/embryology , Heart/growth & development , Myocardium/metabolism , Potassium Channels/biosynthesis , Potassium Channels/genetics , Animals , Cyclic Nucleotide-Gated Cation Channels/biosynthesis , Cyclic Nucleotide-Gated Cation Channels/genetics , Gene Expression , Gene Expression Profiling , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Ion Channels/biosynthesis , Ion Channels/genetics , Mice , Mice, Inbred C57BL , Potassium Channels, Inwardly Rectifying/biosynthesis , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels, Voltage-Gated/biosynthesis , Potassium Channels, Voltage-Gated/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
19.
Neuroscience ; 162(2): 453-61, 2009 Aug 18.
Article in English | MEDLINE | ID: mdl-19409968

ABSTRACT

Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are active at resting membrane potential and thus contribute to neuronal excitability. Their increased activity has recently been demonstrated in models of nerve injury-induced pain. The major aim of the current study was to investigate altered HCN channel protein expression in trigeminal sensory neurons following inflammation of the dura. HCN1 and HCN2 channel immunoreactivity was observed on the membranes of medium- to large-sized trigeminal ganglion neurons with 76% and 85% of HCN1 and HCN2 expressing neurons also containing the 200 kDa neurofilament protein (associated with myelinated fibers). Western immunoblots of lysates from rat trigeminal ganglia also showed bands with appropriate molecular weights for HCN1 and HCN2. Three days after application of complete Freund's adjuvant (CFA) to the dura mater, Western blot band densities were significantly increased; compared to control, to 166% for HCN1 and 284% for HCN2 channel protein. The band densities were normalized against alpha-actin. In addition, the number of retrogradely labeled neurons from the dura expressing HCN1 and HCN2 was significantly increased to 247% (HCN1) and 171% (HCN2), three days after inflammation. When the opioid receptor partial agonist, buprenorphine, was given systemically, immediately after CFA, the inflammation-induced increase in HCN protein expression in both Western blot and immunohistochemical experiments was not observed. These results suggest that HCN1 and HCN2 are involved in inflammation-induced sensory neuron hyperexcitability, and indicate that an opioid receptor agonist can reverse the protein upregulation.


Subject(s)
Analgesics, Opioid/pharmacology , Buprenorphine/pharmacology , Cyclic Nucleotide-Gated Cation Channels/physiology , Ion Channels/physiology , Neurons/metabolism , Potassium Channels/physiology , Trigeminal Ganglion/metabolism , Animals , Brain/blood supply , Brain/metabolism , Cyclic Nucleotide-Gated Cation Channels/biosynthesis , Dura Mater/metabolism , Freund's Adjuvant , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Inflammation/chemically induced , Inflammation/metabolism , Ion Channel Gating , Ion Channels/biosynthesis , Male , Neurofilament Proteins/metabolism , Potassium Channels/biosynthesis , Rats , Rats, Sprague-Dawley , Receptors, Opioid/agonists
20.
J Cardiovasc Pharmacol ; 53(3): 223-30, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19247193

ABSTRACT

G-protein-coupled receptors (GPCRs) represent the largest family of transmembrane receptors involved in cell signal transduction. Many of these GPCRs convey their pharmacological actions by regulating intracellular levels of 3',5'-cyclic adenosine monophosphate (cAMP). Although the heart expresses more than 100 GPCRs, drug agonists for approximately one third of these GPCRs have not been identified. The goal of this project was to initiate the development of a high-throughput screening assay for monitoring cAMP in the heart. Neonatal rat cardiac ventricular myocytes were isolated and cultured on coverslips (whole-cell patch clamp recording) or in 96-well plates (fluorescent imaging plate reader measurements). Cells were infected with adenovirus expressing either beta-galactosidase (AdLacZ) or a mutant cyclic nucleotide-gated (CNG) channel containing the double mutation C460W/E583M (AdCNG). Addition of 2 microM forskolin along with 100 microM 3-isobutyl-1-methylxanthine, to increase intracellular cAMP, activated a cation current in myocytes infected with the AdCNG. In myocytes loaded with the fluorescent Ca indicator Fluo-4, stimulation with forskolin, epinephrine, norepinephrine, or the beta-adrenergic receptor agonist isoproterenol increased the fluorescent signal indicative of Ca influx through the CNG channel. In conclusion, CNG channels are readily expressed in cultured cardiac myocytes and may be utilized in high-throughput screening assays of intracellular cAMP.


Subject(s)
Biosensing Techniques/methods , Cyclic AMP/analysis , Cyclic Nucleotide-Gated Cation Channels/metabolism , Heart Ventricles/metabolism , Myocytes, Cardiac/metabolism , Adenoviridae/enzymology , Adenoviridae/genetics , Aniline Compounds/chemistry , Animals , Animals, Newborn , Cells, Cultured , Cyclic AMP/metabolism , Cyclic Nucleotide-Gated Cation Channels/biosynthesis , Cyclic Nucleotide-Gated Cation Channels/genetics , Fluorescent Dyes/chemistry , Heart Ventricles/cytology , Membrane Potentials/physiology , Patch-Clamp Techniques , Rats , Xanthenes/chemistry , beta-Galactosidase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...