Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Biochem Cell Biol ; 95(5): 531-536, 2017 10.
Article in English | MEDLINE | ID: mdl-28708947

ABSTRACT

The KCNH1 gene encodes the Kv10.1 (Eag1) ion channel, a member of the EAG (ether-à-go-go) family of voltage-gated potassium channels. Recent studies have demonstrated that KCHN1 mutations are implicated in Temple-Baraitser and Zimmermann-Laband syndromes and other forms of developmental deficits that all present with mental retardation and epilepsy, suggesting that Kv10.1 might be important for cognitive development in humans. Although the Kv10.1 channel is mainly expressed in the mammalian brain, its ectopic expression occurs in 70% of human cancers. Cancer cells and tumors expressing Kv10.1 acquire selective advantages that favor cancer progression through molecular mechanisms that involve several cellular pathways, indicating that protein-protein interactions may be important for Kv10.1 influence in cell proliferation and tumorigenesis. Several studies on transcriptional and post-transcriptional regulation of Kv10.1 expression have shown interesting mechanistic insights about Kv10.1 role in oncogenesis, increasing the importance of identifying the cellular factors that regulate Kv10.1 expression in tumors.


Subject(s)
Brain/metabolism , Ether-A-Go-Go Potassium Channels/metabolism , Neoplasms/metabolism , Animals , Humans
2.
Mol Psychiatry ; 19(10): 1143-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-23999527

ABSTRACT

In 2007, a multifaceted syndrome, associated with anti-NMDA receptor autoantibodies (NMDAR-AB) of immunoglobulin-G isotype, has been described, which variably consists of psychosis, epilepsy, cognitive decline and extrapyramidal symptoms. Prevalence and significance of NMDAR-AB in complex neuropsychiatric disease versus health, however, have remained unclear. We tested sera of 2817 subjects (1325 healthy, 1081 schizophrenic, 263 Parkinson and 148 affective-disorder subjects) for presence of NMDAR-AB, conducted a genome-wide genetic association study, comparing AB carriers versus non-carriers, and assessed their influenza AB status. For mechanistic insight and documentation of AB functionality, in vivo experiments involving mice with deficient blood-brain barrier (ApoE(-/-)) and in vitro endocytosis assays in primary cortical neurons were performed. In 10.5% of subjects, NMDAR-AB (NR1 subunit) of any immunoglobulin isotype were detected, with no difference in seroprevalence, titer or in vitro functionality between patients and healthy controls. Administration of extracted human serum to mice influenced basal and MK-801-induced activity in the open field only in ApoE(-/-) mice injected with NMDAR-AB-positive serum but not in respective controls. Seropositive schizophrenic patients with a history of neurotrauma or birth complications, indicating an at least temporarily compromised blood-brain barrier, had more neurological abnormalities than seronegative patients with comparable history. A common genetic variant (rs524991, P=6.15E-08) as well as past influenza A (P=0.024) or B (P=0.006) infection were identified as predisposing factors for NMDAR-AB seropositivity. The >10% overall seroprevalence of NMDAR-AB of both healthy individuals and patients is unexpectedly high. Clinical significance, however, apparently depends on association with past or present perturbations of blood-brain barrier function.


Subject(s)
Autoantibodies/blood , Blood-Brain Barrier/metabolism , Mood Disorders/metabolism , Parkinson Disease/metabolism , Receptors, N-Methyl-D-Aspartate/immunology , Schizophrenia/metabolism , Adult , Aged , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Cerebral Cortex/metabolism , Endocytosis/physiology , Female , Genome-Wide Association Study , Humans , Influenza, Human/genetics , Influenza, Human/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Mood Disorders/genetics , Neurons/metabolism , Parkinson Disease/genetics , Polymorphism, Single Nucleotide , Receptors, N-Methyl-D-Aspartate/genetics , Schizophrenia/genetics
3.
Neurosci Lett ; 559: 13-7, 2014 Jan 24.
Article in English | MEDLINE | ID: mdl-24284010

ABSTRACT

Recently, our group described the ether-à-go-go1(Eag1) voltage-gated potassium (K(+)) channel (Kv10.1) expression in the dopaminergic cells indicating that these channels are part of the diversified group of ion channels related to dopaminergic neurons function. The increase of dopamine neurotransmission induces a reduction in the prepulse inhibition (PPI) of the acoustic startle reflex in rodents, which is a reliable index of sensorimotor gating deficits. The PPI response has been reported to be abnormally reduced in schizophrenia patients. The role of Eag1 K(+) channels in the PPI reaction had not been revealed until now, albeit the singular distribution of Eag1 in the dentate gyrus of the hippocampus and the hippocampal regulation of the startle reflex and PPI. The aim of this work was to investigate if Eag1 blockade on hippocampus modifies the PPI-disruptive effects of apomorphine in Wistar rats. Bilateral injection of anti-Eag1 single-chain antibody into the dentate gyrus of hippocampus did not modify apomorphine-disruptive effects in the PPI response. However, Eag1 antibody completely restored the startle amplitude decrease revealed after dentate gyrus surgery. These potentially biological important phenomenon merits further investigation regarding the role of Eag1 K(+) channels, mainly, on startle reflex modulation, since the physiological role of these channels remain obscure.


Subject(s)
Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ether-A-Go-Go Potassium Channels/physiology , Hippocampus/physiology , Neural Inhibition/physiology , Sensory Gating/physiology , Acoustic Stimulation/methods , Animals , Antibodies, Anti-Idiotypic/pharmacology , Apomorphine/pharmacology , Dopamine Agonists/pharmacology , Hippocampus/drug effects , Male , Neural Inhibition/drug effects , Rats , Rats, Wistar , Sensory Gating/drug effects
4.
Curr Med Chem ; 19(5): 675-82, 2012.
Article in English | MEDLINE | ID: mdl-22204340

ABSTRACT

K(V)10.1 has recently become generally accepted as a promising cancer target, as it is ectopically expressed in the majority of solid tumors. Due to its cell-surface accessibility, K(V)10.1 has a strong potential for tumor treatment and diagnosis. Given that its mode of action is likely independent of conventional cancer pathways such as tyrosine kinases, K(V)10.1 opens a novel window for treating cancer. In this review we will give an overview of the current status of data linking K(V)10.1 to cancer, and propose techniques that could exploit K(V)10.1's properties for the management of cancer.


Subject(s)
Ether-A-Go-Go Potassium Channels/therapeutic use , Neoplasms , Humans , Molecular Targeted Therapy , Neoplasms/diagnosis , Neoplasms/therapy
5.
J Neurosci Res ; 90(3): 632-40, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22006722

ABSTRACT

Transient global brain ischemia causes delayed neuronal death in the hippocampus that has been associated with impairments in hippocampus-dependent brain function, such as mood, learning, and memory. We investigated the expression of voltage-dependent Kcnh1 and Kcnh5, ether à go-go-related Eag1 and Eag2 (K(V) 10.1 and K(V) 10.2), and small-conductance calcium-activated SK3 (K(Ca) 2.3, Kcnn3) K(+) channels in the hippocampus in rats after transient global brain ischemia. We tested whether the expression of these channels is associated with behavioral changes by evaluating the animals in the elevated plus maze and step-down inhibitory avoidance task. Seven or tweny-eight days after transient global brain ischemia, one group of rats had the hippocampus bilaterally dissected, and mRNA levels were determined. Seven days after transient global brain ischemia, the rats exhibited a decrease in anxiety-like behavior and memory impairments. An increase in anxiety levels was detected 28 days after ischemia. Eag2 mRNA downregulation was observed in the hippocampus 7 days after transient global brain ischemia, whereas Eag1 and SK3 mRNA expression remained unaltered. This is the first experimental evidence that transient global brain ischemia temporarily alters Eag2. The number of intact-appearing pyramidal neurons was substantially decreased in CA1 and statistically measurable in CA2, CA3, and CA4 hippocampal subfields compared with sham control animals 7 or 28 days after ischemia. mRNA expression in the rat hippocampus. The present results provide further information for the characterization of the physiological role of Eag2 channels in the central nervous system.


Subject(s)
Brain Ischemia/metabolism , Ether-A-Go-Go Potassium Channels/metabolism , Hippocampus/metabolism , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Animals , Avoidance Learning/physiology , Behavior, Animal/physiology , Brain Ischemia/physiopathology , Down-Regulation , Hippocampus/physiopathology , Male , Maze Learning/physiology , Neurons/metabolism , Rats , Rats, Wistar
6.
Neurotox Res ; 21(3): 317-33, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22048886

ABSTRACT

The ether à go-go (Eag) gene encodes the voltage-gated potassium (K(+)) ion channel Kv10.1, whose function still remains unknown. As dopamine may directly affect K(+) channels, we evaluated whether a nigrostriatal dopaminergic lesion induced by the neurotoxin 6-hydroxydopamine (6-OHDA) would alter Eag1-K(+) channel expression in the rat basal ganglia and related brain regions. Male Wistar rats received a microinjection of either saline or 6-OHDA (unilaterally) into the medial forebrain bundle. The extent of the dopaminergic lesion induced by 6-OHDA was evaluated by apomorphine-induced rotational behavior and by tyrosine hydroxylase (TH) immunoreactivity. The 6-OHDA microinjection caused a partial or complete lesion of dopaminergic cells, as well as a reduction of Eag1+ cells in a manner proportional to the extent of the lesion. In addition, we observed a decrease in TH immunoreactivity in the ipsilateral striatum. In conclusion, the expression of the Eag1-K(+)-channel throughout the nigrostriatal pathway in the rat brain, its co-localization with dopaminergic cells and its reduction mirroring the extent of the lesion highlight a physiological circuitry where the functional role of this channel can be investigated. The Eag1-K(+) channel expression in dopaminergic cells suggests that these channels are part of the diversified group of ion channels that generate and maintain the electrophysiological activity pattern of dopaminergic midbrain neurons.


Subject(s)
Basal Ganglia/metabolism , Dopaminergic Neurons/metabolism , Ether-A-Go-Go Potassium Channels/metabolism , Nerve Degeneration/metabolism , Oxidopamine/toxicity , Parkinsonian Disorders/metabolism , Animals , Basal Ganglia/drug effects , Basal Ganglia/pathology , Disease Models, Animal , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Globus Pallidus/drug effects , Globus Pallidus/metabolism , Globus Pallidus/pathology , Immunohistochemistry , Male , Nerve Degeneration/chemically induced , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Nucleus Accumbens/pathology , Parkinsonian Disorders/chemically induced , Rats , Rats, Wistar , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Substantia Nigra/pathology , Sympatholytics/toxicity , Tyrosine 3-Monooxygenase/metabolism
7.
Neuroscience ; 155(3): 833-44, 2008 Aug 26.
Article in English | MEDLINE | ID: mdl-18650019

ABSTRACT

Eag1 (K(V)10.1) is the founding member of an evolutionarily conserved superfamily of voltage-gated K(+) channels. In rats and humans Eag1 is preferentially expressed in adult brain but its regional distribution has only been studied at mRNA level and only in the rat at high resolution. The main aim of the present study is to describe the distribution of Eag1 protein in adult rat brain in comparison to selected regions of the human adult brain. The distribution of Eag1 protein was assessed using alkaline-phosphatase based immunohistochemistry. Eag1 immunoreactivity was widespread, although selective, throughout rat brain, especially noticeable in the perinuclear space of cells and proximal regions of the extensions, both in rat and human brain. To relate the results to the relative abundance of Eag1 transcripts in different regions of rat brain a reverse-transcription coupled to quantitative polymerase chain reaction (real time PCR) was performed. This real time PCR analysis showed high Eag1 expression in the olfactory bulb, cerebral cortex, hippocampus, hypothalamus, and cerebellum. The results indicate that Eag1 protein expression greatly overlaps with mRNA distribution in rats and humans. The physiological relevance of potassium channels in the different regions expressing Eag1 protein is discussed.


Subject(s)
Central Nervous System/metabolism , Ether-A-Go-Go Potassium Channels/metabolism , Adult , Aged , Animals , Central Nervous System/anatomy & histology , Ether-A-Go-Go Potassium Channels/genetics , Female , Humans , Male , Middle Aged , RNA, Messenger/metabolism , Rats , Rats, Wistar
8.
J Membr Biol ; 205(3): 115-24, 2005 Jun.
Article in English | MEDLINE | ID: mdl-16362499

ABSTRACT

Ion channels are being associated with a growing number of diseases including cancer. This overview summarizes data on voltage-gated potassium channels (VGKCs) that exhibit oncogenic properties: ether-à-go-go type 1 (Eag1). Normally, Eag1 is expressed almost exclusively in tissue of neural origin, but its ectopic expression leads to uncontrolled proliferation, while inhibition of Eag1 expression produces a concomitant reduction in proliferation. Specific monoclonal antibodies against Eag1 recognize an epitope in over 80% of human tumors of diverse origins, endowing it with diagnostic and therapeutic potential. Eag1 also possesses unique electrophysiological properties that simplify its identification. This is particularly important, as specific blockers of Eag1 currents are not available. Molecular imaging of Eag1 in live tumor models has been accomplished with dye-tagged antibodies using 3-D imaging techniques in the near-infrared spectral range.


Subject(s)
Neoplasms/etiology , Potassium Channels, Voltage-Gated/physiology , Animals , Cell Proliferation , Cell Size , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ether-A-Go-Go Potassium Channels/physiology , Humans , Membrane Potentials , Neoplasms/drug therapy , Neoplasms/physiopathology , Potassium Channel Blockers/therapeutic use
9.
J Dairy Sci ; 86(1): 52-9, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12613848

ABSTRACT

Several lines of research support a role for human milk oligosaccharides in the defense of breast-fed infants against pathogens. Some ofthese oligosaccharides contain at least one moiety of sialic acid and are, thus, termed sialyloligosaccharides. These constitute a significant component (>1 g/L) of human milk. It is well established that milk composition varies among species, and previous reports have indicated that one ofthe differences between human and bovine milk is precisely their contents of sialyloligosaccharides. Because most infant formulas are manufactured with bovine milk components, it follows that formula-fed and breast-fed infants ingest dissimilar quantities of these carbohydrate structures. To ascertain these differences and their impact along lactation, the contents of oligosaccharide-bound sialic acids and major sialyloligosaccharides in samples of human and bovine milk (obtained at different lactation stages) were determined. In addition, infant formulas were assayed for their sialyloligosaccharide contents. Seven sialyloligosaccharides were identified in human milk; namely, 3'-sialyl-3-fucosyllactose and sialyllacto-N-tetraoses (a and b+c), the predominant structures at all lactation stages. Five sialyloligosaccharides were identified in bovine milk, of which 6'-sialyllactosamine and 3'-sialyllactose were the most abundant. In addition, sialyloligosaccharides in human and bovine milk decreased along lactation, and infant formulas did not contain significant amounts of sialyloligosaccharides. The results point to the general conclusion that regarding both qualitative and quantitative aspects, milk from humans and cows and infant formulas have different oligosaccharide contents. In this sense, bottle-fed infants are subject to reduced sialyloligosaccharide intake as compared to breast-fed infants.


Subject(s)
Cattle/physiology , Infant Food/analysis , Lactation/metabolism , Milk, Human/chemistry , Milk/chemistry , Oligosaccharides/analysis , Animal Nutritional Physiological Phenomena , Animals , Animals, Suckling , Cattle/metabolism , Chromatography, High Pressure Liquid , Female , Humans , Infant , Infant Nutritional Physiological Phenomena , Infant, Newborn , Lactation/physiology , Neuraminidase/metabolism , Oligosaccharides/chemistry , Species Specificity
11.
J Dairy Sci ; 84(5): 995-1000, 2001 May.
Article in English | MEDLINE | ID: mdl-11384055

ABSTRACT

The sialoglycoconjugate content of human milk has been extensively studied. However, little attention has been paid to the changes occurring in these compounds in bovine milk during lactation. Since sialoglycoconjugates are very abundant in milk from the early stages of lactation, they have been suggested to be important for the nutrition of the newborn during the first months of life. The distribution of sialoglycoconjugates (expressed as glycoconjugate-bound sialic acid) from four different stages of lactation (colostrum, transitional, mature, and late-lactation milks) was investigated in four Spanish-Brown cows. All the fractions studied (total sialic acids, glycoproteins, oligosaccharides, casein, and gangliosides) showed a similar trend. We found the highest values in the colostrum, these decreasing in transitional and mature milks and increasing again in late-lactation milk. We also found a selective change in the relative contents of glycoprotein- and oligosaccharide-bound sialic acids. In mature milk, the latter increased up to 80% (59% in colostrum) and the former decreased to 3.9% (35.3% in colostrum). It would appear that the decrease in oligosaccharide-bound sialic acid is compensated by the increase in glycoprotein-bound sialic acid. From these results, it is deduced that newborn infants or calves fed with infant formulas or milk replacers, respectively, should be supplemented with sialoglycoconjugates to approximate the composition of human and cow milk as far as is practicable.


Subject(s)
Animals, Suckling/metabolism , Cattle/physiology , Colostrum/chemistry , Gangliosides/analysis , Milk/chemistry , Animal Nutritional Physiological Phenomena , Animals , Female , Glycoproteins/analysis , Lactation , Oligosaccharides/analysis
12.
Pflugers Arch ; 441(2-3): 167-74, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11211100

ABSTRACT

The electrophysiological properties of ether a go-go (EAG) potassium channels are modified during the cell cycle when they are expressed in heterologous systems. In Chinese hamster ovary (CHO) mammalian somatic cells we found that the cell-cycle-dependent modulation of human EAG (hEAG) channels occurs during the M phase. This modulation has three components: reduction in current density, increased sensitivity to block by intracellular sodium, and increased selectivity for potassium ions. In this work, these three properties have been used to define the mitotic phenotype of EAG currents. The signaling pathway leading to such changes of channel properties is unknown. We tested the hypothesis that cytoskeletal interactions might affect the electrophysiological changes observed during the cell cycle. The disruption of actin filaments induces a significant increase in current density, without inducing the cell-cycle-related phenotype. In contrast, disturbance of the microtubules, achieved by pharmacological means or by mechanical excision of the membrane patch, does induce the cell-cycle-related phenotype. Our results demonstrate that hEAG channels establish complex interactions with cytoskeletal elements, and that these interactions strongly influence the properties of the channels. We also conclude that the electrophysiological changes observed during the cell cycle are most likely due to reorganization of the cytoskeleton during the G2/M transition.


Subject(s)
Cytoskeleton/physiology , Potassium Channels/physiology , Actin Cytoskeleton/physiology , Actins/physiology , Animals , CHO Cells , Cell Cycle , Cesium/metabolism , Cricetinae , Electric Conductivity , Electrophysiology , Ether-A-Go-Go Potassium Channels , Humans , Microtubules/physiology , Mitosis , Potassium/metabolism , Signal Transduction , Sodium/pharmacology
13.
EMBO J ; 18(20): 5540-7, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10523298

ABSTRACT

We have investigated the possible implication of the cell cycle-regulated K(+) channel ether à go-go (EAG) in cell proliferation and transformation. We show that transfection of EAG into mammalian cells confers a transformed phenotype. In addition, human EAG mRNA is detected in several somatic cancer cell lines, despite being preferentially expressed in brain among normal tissues. Inhibition of EAG expression in several of these cancer cell lines causes a significant reduction of cell proliferation. Moreover, the expression of EAG favours tumour progression when transfected cells are injected into immune-depressed mice. These data provide evidence for the oncogenic potential of EAG.


Subject(s)
Cell Transformation, Neoplastic , Potassium Channels/physiology , Animals , Base Sequence , Cell Division/drug effects , Cell Division/genetics , Cell Division/physiology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cloning, Molecular , DNA Primers/genetics , Ether-A-Go-Go Potassium Channels , Gene Expression , Humans , In Situ Hybridization, Fluorescence , Mice , Mice, SCID , Molecular Sequence Data , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Oligodeoxyribonucleotides, Antisense/genetics , Oligodeoxyribonucleotides, Antisense/pharmacology , Phenotype , Potassium Channels/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Cells, Cultured
14.
J Cell Biol ; 143(3): 767-75, 1998 Nov 02.
Article in English | MEDLINE | ID: mdl-9813096

ABSTRACT

Release from arrest in G2 phase of the cell cycle causes profound changes in rat ether-à-go-go (r-eag) K+ channels heterologously expressed in Xenopus oocytes. The most evident consequence of the onset of maturation is the appearance of rectification in the r-eag current. The trigger for these changes is located downstream of the activation of mitosis-promoting factor (MPF). We demonstrate here that the rectification is due to a voltage-dependent block by intracellular Na+ ions. Manipulation of the intracellular Na+ concentration indicates that the site of Na+ block is located approximately 45% into the electrical distance of the pore and is only present in oocytes undergoing maturation. Since the currents through excised patches from immature oocytes exhibited a fast rundown, we studied CHO-K1 cells permanently transfected with r-eag. These cells displayed currents with a variable degree of block by Na+ and variable permeability to Cs+. Partial synchronization of the cultures in G0/G1 or M phases of the cell cycle greatly reduced the variability. The combined data obtained from mammalian cells and oocytes strongly suggest that the permeability properties of r-eag K+ channels are modulated during cell cycle-related processes.


Subject(s)
Cell Cycle/physiology , Potassium Channels/physiology , Animals , CHO Cells , Cell Membrane Permeability , Cesium/metabolism , Cricetinae , Enzyme Activation , Ether-A-Go-Go Potassium Channels , Intracellular Fluid/physiology , Ions , Maturation-Promoting Factor/metabolism , Sodium/metabolism
15.
Pflugers Arch ; 434(6): 661-8, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9305996

ABSTRACT

Members of the rat brain Kv1 family of cloned potassium channels are structurally highly homologous, but have diverse conductance and pharmacological characteristics. Here we present data on the effects of mutating residues K533 in the P-region and H471 in the S4-S5 linker of Kv1.4 to their equivalent residues in Kv1.1 and Kv1.6 on single-channel conductance and sensitivity to external tetraethylammonium cations (TEA+) and internal Mg2+. Exchange of residue K533 for its equivalent residue (Y) in Kv1.1 and Kv1.6 increased the single-channel conductance at both negative and positive potentials. This mutation is known to reduce the IC50 for external TEA+ from > 100 mM to 0.6 mM, almost identical to that for Kv1.1 (0.53 mM). We have now found that the additional exchange of residue H471 for the equivalent residue (K) in Kv1.6 increased the IC50 for external TEA+ from 0.6 mM (Kv1.4K533Y) to 2.39 mM; this is very close to that for wild-type Kv1.6 channels (2.84 mM). The mutation H471K alone was ineffective. We thus provide evidence that the S4-S5 linker does contribute to the channel's inner-pore region. Data on the block of Kv1 channels by internal Mg2+ indicate that while the binding site is probably situated within the deep-pore region, its exact location may be channel specific.


Subject(s)
Brain/metabolism , Molecular Biology/methods , Multigene Family/genetics , Potassium Channels/genetics , Potassium Channels/metabolism , Animals , Electric Conductivity , Female , Magnesium/pharmacology , Mutation/genetics , Potassium Channels/physiology , Rats , Tetraethylammonium/pharmacology , Xenopus laevis
16.
Biochemistry ; 36(11): 3308-18, 1997 Mar 18.
Article in English | MEDLINE | ID: mdl-9116009

ABSTRACT

Glutamine 105 in the third transmembrane helix of the thyrotropin-releasing hormone receptor (TRH-R) occupies a position equivalent to a conserved negatively charged residue in receptors for biogenic amines where it acts as counterion interacting with the cationic amine moiety of the ligand. Maximum levels of response to TRH in oocytes expressing wild-type TRH-Rs were indistinguishable from those of oocytes expressing receptors mutated to Glu, Asn, or Asp in position 105. However, the EC50 values for activation of oocyte responses increased more than 500 times in oocytes expressing mutant Glu105 receptors, in which the amido group of Gln105 has been removed by site-directed mutagenesis. Charge effects do not seem to be involved in the huge effect of mutating Gln105 to Glu, since mutation of Gln105 to Asp induces only a 15-fold increase in EC50. Furthermore, no change in EC50 is observed after mutation of Asn110 to Asp. The affinity shift (identified by changes in EC50 values for systems of comparable efficacy) in Glu105 mutant receptors was partially recovered in oocytes expressing Asn105 mutant receptors. These results and those obtained after substitution of Lys, Leu, Tyr, and Ser for Gln105 suggest that the presence and the correct position of the Gln hydrogen bond-donor amido group are important for normal functionality of the receptor. In wild type or Asp105 mutant receptors showing the same maximal responses, decreases in affinity with TRH and methyl-histidyl-TRH correlated with increased dissociation rates of hormone from the receptor. Rapid dilution experiments following subsecond stimulation indicate that the TRH-R is converted rapidly from a form showing fast dissociation kinetics to a form from which the hormone dissociates slowly. Mutation of residue 105 impairs the receptor shift between these two forms. This effect was demonstrated in a direct way by comparing [3H]methyl-histidyl-TRH dissociation rates in COS-7 cells transfected with either wild type or Asp105 mutant TRH-Rs. Thus, residues located in transmembrane helix III positions equivalent to those of the counterions for biogenic amines, regulate hormone-receptor interactions in the TRH receptor (and perhaps other receptors). Furthermore, the nature of the amino acid in these positions may also play a role, directly or indirectly, in conformational changes leading to receptor activation, and hence to signal transduction.


Subject(s)
Glutamine , Point Mutation , Protein Structure, Secondary , Receptors, Cell Surface/chemistry , Receptors, Thyrotropin-Releasing Hormone/chemistry , Receptors, Thyrotropin-Releasing Hormone/metabolism , Thyrotropin-Releasing Hormone/chemistry , Amino Acid Sequence , Animals , Aspartic Acid , COS Cells , Conserved Sequence , Female , Hormones/chemistry , Kinetics , Ligands , Molecular Sequence Data , Mutagenesis, Site-Directed , Oocytes/physiology , Receptors, Thyrotropin-Releasing Hormone/physiology , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Thyrotropin-Releasing Hormone/metabolism , Thyrotropin-Releasing Hormone/pharmacology , Transfection
17.
Proc Natl Acad Sci U S A ; 94(2): 537-42, 1997 Jan 21.
Article in English | MEDLINE | ID: mdl-9012819

ABSTRACT

The cell cycle is the crucial process that leads to mitosis in all cell types. The dramatic redirectioning of many cellular processes during the cycle is known to involve ion channels, either changing their level of expression or their voltage dependence, as in the case of inward rectifiers. Here we describe the specific inhibition of heterologously expressed ionic channels at the onset of maturation in Xenopus oocytes. In cells expressing rat eag (R-eag) potassium channels, maturation induces a dramatic reduction in the current amplitude, which is almost complete in most cases. The key molecule in oocyte maturation, the mitosis-promoting factor (a complex of cyclin B and p34cdc2), is able to induce similar changes when injected into the oocytes.


Subject(s)
Maturation-Promoting Factor/physiology , Potassium Channels/physiology , Animals , Cyclins/physiology , Electric Conductivity , Ether-A-Go-Go Potassium Channels , Ion Channel Gating , Membrane Potentials , Oocytes , Progesterone/pharmacology , Signal Transduction , Spermine/pharmacology , Time Factors , Xenopus laevis
18.
Pflugers Arch ; 435(1): 119-29, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9359911

ABSTRACT

Reduction of an inwardly rectifying K+ current by thyrotropin-releasing hormone (TRH) and caffeine has been considered to be an important determinant of electrical activity increases in GH3 rat anterior pituitary cells. However, the existence of an inwardly rectifying K+ current component was recently regarded as a misidentification of an M-like outward current, proposed to be the TRH target in pituitary cells, including GH3 cells. In this report, an inwardly rectifying component of K+ current is indeed demonstrated in perforated-patch voltage-clamped GH3 cells. The degree of rectification varied from cell to cell, but both TRH and caffeine specifically blocked a fraction of current with strong rectification in the hyperpolarizing direction. Use of ramp pulses to continuously modify the membrane potential demonstrated a prominent blockade even in cells with no current reduction at voltages at which M-currents are active. Depolarization steps to positive voltages at the maximum of the inward current induced a caffeine-sensitive instantaneous outward current followed by a single exponential decay. The magnitude of this current was modified in a biphasic way according to the duration of the previous hyperpolarization step. The kinetic characteristics of the current are compatible with the possibility that removal from inactivation of a fast-inactivating delayed rectifier causes the hyperpolarization-induced current. Furthermore, the inwardly rectifying current was blocked by astemizole, a potent and selective inhibitor of human ether-á-go-go -related gene (HERG) K+ channels. Along with other pharmacological and kinetic evidence, this indicates that the secretagogue-regulated current is probably mediated by a HERG-like K+ channel. Addition of astemizole to current-clamped cells induced clear increases in the frequency of action potential production. Thus, an inwardly-rectifying K+ current and not an M-like outward current seems to be involved in TRH and caffeine modulation of electrical activity in GH3 cells.


Subject(s)
Caffeine/pharmacology , Cation Transport Proteins , DNA-Binding Proteins , Pituitary Gland, Anterior/physiology , Potassium Channels, Voltage-Gated , Potassium Channels/drug effects , Potassium Channels/physiology , Thyrotropin-Releasing Hormone/pharmacology , Trans-Activators , Action Potentials/drug effects , Animals , Astemizole/pharmacology , Calcium/pharmacology , Cell Line , ERG1 Potassium Channel , Electric Conductivity , Ether-A-Go-Go Potassium Channels , Humans , Kinetics , Membrane Potentials , Pituitary Gland, Anterior/drug effects , Rats , Transcriptional Regulator ERG
19.
Article in English | MEDLINE | ID: mdl-9417989

ABSTRACT

The activity of UDP-N-acetylglucosamine 2-epimerase was determined in the liver of rats and guinea-pigs of different ages. The activity of this enzyme in rats was low at birth, increased to a maximum value on day 15, and fell gradually until day 30. Thereafter, it increased up to the 60th day. The activity profile of the enzyme from guinea-pig liver was very similar. However, guinea-pig activity was 2-5 times lower than in rats. Both rats and guinea-pigs displayed similar liver sialic acid contents which increased from birth to 2 months of age. Rats also showed a N-glycolylneuraminic acid content that decreased from birth to 2 months. From these results we can inferred that postnatal UDP-N-acetylglucosamine 2-epimerase activity seems to be correlated with age and the developmental states of rats and guinea-pigs.


Subject(s)
Escherichia coli Proteins , Liver/enzymology , Liver/growth & development , Animals , Animals, Newborn , Carbohydrate Epimerases/metabolism , Female , Guinea Pigs , Liver/metabolism , Male , Milk/metabolism , N-Acetylneuraminic Acid/metabolism , Neuraminic Acids/metabolism , Rats , Rats, Wistar , Species Specificity
20.
Pflugers Arch ; 431(3): 443-51, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8584440

ABSTRACT

Treatment of rat anterior pituitary GH3 cells with caffeine causes a reversible enhancement of electrical activity superimposed over a depolarization of the plasma membrane potential. Similar results are obtained with theophylline, but not with isobutylmethylxanthine or forskolin. The effects of caffeine are not related to Ca2+ liberation from intracellular stores since they are not affected by incubation of the cells with ryanodine or thapsigargin. Furthermore, caffeine-induced hyperpolarization of the membrane is not detectable even in cells in which Ca2+ liberation from inositol 1,4,5-trisphosphate-sensitive compartments produces a prominent transient hyperpolarization in response to thyrotropin-releasing hormone. Reductions of Ca2+-dependent K+ currents caused by partial block of L-type Ca2+ channels by caffeine are not sufficient to explain the effects of the xanthine, since the results obtained with caffeine are not mimicked by direct blockade of Ca2+ channels with nisoldipine. GH3 cell inwardly rectifying K+ currents are inhibited by caffeine. Studies on the voltage dependence of the caffeine-induced effects indicate a close correlation between alterations of electrical parameters and reported values of steady-state voltage dependence of inactivation of these currents. We conclude that, as previously shown for thyrotropin-releasing hormone, modulation of inwardly rectifying K+ currents plays a major role determining the firing rate of GH3 cells and its enhancement by caffeine.


Subject(s)
Caffeine/pharmacology , Pituitary Gland, Anterior/physiology , Potassium Channels/drug effects , Potassium Channels/physiology , Animals , Calcium/metabolism , Calcium Channels/drug effects , Cell Line , Membrane Potentials/drug effects , Pituitary Gland, Anterior/cytology , Pituitary Gland, Anterior/drug effects , Rats , Terpenes/pharmacology , Thapsigargin
SELECTION OF CITATIONS
SEARCH DETAIL
...