Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
FEBS Lett ; 594(17): 2914-2922, 2020 09.
Article in English | MEDLINE | ID: mdl-32484574

ABSTRACT

Cav2.2 N-type voltage-dependent Ca2+ channel (VDCC) expressed in neurons is known to be essential for neurotransmitter release. We have shown previously that this channel is also expressed in nonexcitable microglia and plays pivotal roles in microglial functions. Here, we have examined the effects of microglia-specific knockdown (KD) of Cav2.2 channel in a mouse model of Parkinson's disease (PD). We found that the KD of Cav2.2 channel reduces the accumulation of microglia in the substantia nigra and ameliorates the behavioral deficits in PD model mice. These results are in marked contrast with those found in microglia-specific KD of Cav1.2 L-type channel, where exacerbated symptoms are observed. Our results suggest that blockade of microglial Cav2.2 N-type VDCC is beneficial for the treatment of PD.


Subject(s)
Calcium Channels, N-Type/genetics , Dopaminergic Neurons/metabolism , Microglia/metabolism , Parkinsonian Disorders/genetics , Substantia Nigra/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/administration & dosage , Animals , Calcium Channels, L-Type/deficiency , Calcium Channels, L-Type/genetics , Calcium Channels, N-Type/deficiency , Cell Count , Cell Death/genetics , Dopaminergic Neurons/pathology , Gene Expression , Male , Mice , Mice, Inbred C57BL , Microglia/pathology , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/pathology , Psychomotor Performance/physiology , Selective Estrogen Receptor Modulators/pharmacology , Substantia Nigra/pathology , Tamoxifen/pharmacology
2.
Cell Calcium ; 82: 102059, 2019 09.
Article in English | MEDLINE | ID: mdl-31377554

ABSTRACT

Voltage-dependent calcium channel (VDCC) is generally believed to be active only in excitable cells. However, we have reported recently that N-type VDCC (Cav2.2) could become functional in non-excitable cells under pathological conditions. In the present study, we show that Cav2.2 channels are also functional in physiological microglial activation process. By using a mouse microglial cell line (MG6), we examined the effects of a Cav2.2 blocker on the activation of MG6 cells, when treated with lipopolysaccharide (LPS) / interferon γ (IFNγ) or with interleukin-4 (IL-4). As a result, blocking the activation of Cav2.2 enhanced so-called alternative activation process of microglia (transition to neuroprotective M2 microglia) without changing the efficacy of the transition to neuroinflammatory M1 microglia. This enhanced M2 transition involved the activation of a transcription factor hypoxia inducible factor 2 (HIF-2), since a specific blocker of HIF-2 completely abolished this enhancement. We then examined whether Cav2.2 activation was involved in aging-related neuroinflammation. Using primary culture of microglia, we found that the efficacy of microglial M1 transition was enhanced but that M2 transition was reduced by aging, in agreement with a general notion that aging induces enhanced neuroinflammation. Finally, we show here that the moderate blockade of Cav2.2 expression in microglia restores this age-dependent reduction of microglial M2 transition and reduces the aging-induced exaggerated cytokine response, as revealed by a fast recovery from depressive-like behaviors in microglia-specific Cav2.2 deficient mice. These results suggest a critical role for microglial Cav2.2 channel in the aging-related neuroinflammation.


Subject(s)
Aging/physiology , Calcium Channels, N-Type/metabolism , Microglia/physiology , Neurogenic Inflammation/metabolism , Animals , Cell Line , Immunity , Interferon-gamma/metabolism , Interleukin-4/metabolism , Lipopolysaccharides/immunology , Mice , Mice, Inbred C57BL , Neuroprotection , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism
3.
Sci Rep ; 9(1): 9138, 2019 06 24.
Article in English | MEDLINE | ID: mdl-31235768

ABSTRACT

Cav1.2 channels are an L-type voltage-dependent Ca2+ channel, which is specifically blocked by calcium antagonists. Voltage-dependent Ca2+ channels are generally considered to be functional only in excitable cells like neurons and muscle cells, but recently they have been reported to also be functional in non-excitable cells like microglia, which are key players in the innate immune system and have been shown to be involved in the pathophysiology of Parkinson's disease. Here, we show that Cav1.2 channels are expressed in microglia, and that calcium antagonists enhanced the neuroinflammatory M1 transition and inhibited neuroprotective M2 transition of microglia in vitro. Moreover, intensive degeneration of dopaminergic neurons and accompanying behavioural deficits were observed in microglia-specific Cav1.2 knockdown mice intoxicated with MPTP, a neurotoxin that induces Parkinson's disease-like symptoms, suggesting detrimental effects of microglial Cav1.2 blockade on Parkinson's disease. Therefore, microglial Cav1.2 channel may have neuroprotective roles under physiological conditions and may also contribute to recovery from disease conditions.


Subject(s)
Calcium Channels, L-Type/deficiency , Calcium Channels, L-Type/genetics , Gene Knockdown Techniques , Microglia/metabolism , Parkinson Disease/genetics , Parkinson Disease/pathology , Animals , Biomarkers/metabolism , Disease Models, Animal , Dopaminergic Neurons/pathology , Mice , Microglia/pathology , Parkinson Disease/metabolism
4.
Biochem Biophys Res Commun ; 499(2): 253-259, 2018 05 05.
Article in English | MEDLINE | ID: mdl-29567475

ABSTRACT

Peripheral nerve injury induces neuropathic pain, which is characterized by the tactile allodynia and thermal hyperalgesia. N-type voltage-dependent Ca2+ channel (VDCC) plays pivotal roles in the development of neuropathic pain, since mice lacking Cav2.2, the pore-forming subunit of N-type VDCC, show greatly reduced symptoms of both tactile allodynia and thermal hyperalgesia. Our study on gene expression profiles of the wild-type and N-type VDCC knockout (KO) spinal cord and several pain-related brain regions after spinal nerve ligation (SNL) injury revealed altered expression of genes encoding catalytic subunits of phosphatidylinositol-3 kinase (PI3K). PI3K/Akt/mammalian target of rapamycin (PI3K/Akt/mTOR) signaling is considered to be very important for cancer development and drugs targeting the molecules in this pathway have been tested in oncology trials. In the present study, we have tested whether the changes in expression of molecules in this pathway in mice having spinal nerve injury are causally related to neuropathic pain. Our results suggest that spinal nerve injury induces activation of N-type VDCC and the following Ca2+ entry through this channel may change the expression of genes encoding PI3K catalytic subunits (p110α and p110γ), Akt, retinoid X receptor α (RXRα) and RXRγ. Furthermore, the blockers of the molecules in this pathway are found to be effective in reducing neuropathic pain both at the spinal and at the supraspinal levels. Thus, the activation of PI3K/Akt/mTOR/peroxisome proliferator activated receptor gamma (PPARγ) pathway would be a hallmark of the induction and maintenance of neuropathic pain.


Subject(s)
Neuralgia/metabolism , PPAR gamma/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Analgesics/pharmacology , Animals , Chromones/pharmacology , Disease Models, Animal , Hyperalgesia/complications , Hyperalgesia/drug therapy , Ligation , Mice , Morpholines/pharmacology , Neuralgia/pathology , Oligonucleotide Array Sequence Analysis , Protein Kinase Inhibitors/pharmacology , Ribonucleosides/pharmacology , Ribonucleosides/therapeutic use , Sirolimus/pharmacology , Sirolimus/therapeutic use , Spinal Nerves/drug effects , Spinal Nerves/pathology
5.
Biochem Biophys Res Commun ; 450(1): 142-7, 2014 Jul 18.
Article in English | MEDLINE | ID: mdl-24887565

ABSTRACT

Peripheral nerve injury induces neuropathic pain which is characterized by tactile allodynia and thermal hyperalgesia. N-type voltage-dependent Ca(2+) channel (VDCC) plays pivotal roles in the development of neuropathic pain, since mice lacking Cav2.2, the pore-forming subunit of N-type VDCC, show greatly reduced symptoms of both tactile allodynia and thermal hyperalgesia. Our study on gene expression profiles of the Cav2.2 knockout (KO) spinal cord after spinal nerve ligation (SNL)-injury revealed altered expression of genes known to be expressed in microglia, raising an odd idea that N-type VDCC may function in not only excitable (neurons) but also non-excitable (microglia) cells in neuropathic pain state. In the present study, we have tested this idea by using a transgenic mouse line, in which suppression of Cav2.2 expression can be achieved specifically in microglia/macrophage by the application of tamoxifen. We found SNL-operated transgenic mice exhibited greatly reduced signs of tactile allodynia, whereas the degree of thermal hyperalgesia was almost the same as that of control. Immunohistochemical analysis of the transgenic lumbar spinal cord revealed reduced accumulation of Iba1-positive cells (microglia/macrophage) around the injured neurons, indicating microglial N-type VDCC is important for accumulation of microglia at the lesion sites. Although the mechanism of its activation is not clear at present, activation of N-type VDCC expressed in non-excitable microglial cells contributes to the pathophysiology of neuropathic pain.


Subject(s)
Calcium Channels, N-Type/metabolism , Microglia/metabolism , Microglia/pathology , Neuralgia/pathology , Neuralgia/physiopathology , Spinal Nerves/pathology , Spinal Nerves/physiopathology , Animals , Female , Male , Mice , Mice, Transgenic , Pain Measurement
6.
Mol Pain ; 5: 74, 2009 Dec 18.
Article in English | MEDLINE | ID: mdl-20021638

ABSTRACT

BACKGROUND: Neuropathic pain is a complex chronic pain generated by damage to, or pathological changes in the somatosensory nervous system. Characteristic features of neuropathic pain are allodynia, hyperalgesia and spontaneous pain. Such abnormalities associated with neuropathic pain state remain to be a significant clinical problem. However, the neuronal mechanisms underlying the pathogenesis of neuropathic pain are complex and still poorly understood. Casein kinase 1 is a serine/threonine protein kinase and has been implicated in a wide range of signaling activities such as cell differentiation, proliferation, apoptosis, circadian rhythms and membrane transport. In mammals, the CK1 family consists of seven members (alpha, beta, gamma1, gamma2, gamma3, delta, and epsilon) with a highly conserved kinase domain and divergent amino- and carboxy-termini. RESULTS: Preliminary cDNA microarray analysis revealed that the expression of the casein kinase 1 epsilon (CK1epsilon) mRNA in the spinal cord of the neuropathic pain-resistant N- type Ca2+ channel deficient (Cav2.2-/-) mice was decreased by the spinal nerve injury. The same injury exerted no effects on the expression of CK1epsilon mRNA in the wild-type mice. Western blot analysis of the spinal cord identified the downregulation of CK1epsilon protein in the injured Cav2.2-/- mice, which is consistent with the data of microarray analysis. However, the expression of CK1epsilon protein was found to be up-regulated in the spinal cord of injured wild-type mice. Immunocytochemical analysis revealed that the spinal nerve injury changed the expression profiles of CK1epsilon protein in the dorsal root ganglion (DRG) and the spinal cord neurons. Both the percentage of CK1epsilon-positive neurons and the expression level of CK1epsilon protein were increased in DRG and the spinal cord of the neuropathic mice. These changes were reversed in the spinal cord of the injured Cav2.2-/- mice. Furthermore, intrathecal administration of a CK1 inhibitor IC261 produced marked anti-allodynic and anti-hyperalgesic effects on the neuropathic mice. In addition, primary afferent fiber-evoked spinal excitatory responses in the neuropathic mice were reduced by IC261. CONCLUSIONS: These results suggest that CK1epsilon plays important physiological roles in neuropathic pain signaling. Therefore CK1epsilon is a useful target for analgesic drug development.


Subject(s)
Casein Kinase 1 epsilon/metabolism , Ganglia, Spinal/enzymology , Peripheral Nervous System Diseases/enzymology , Spinal Cord/enzymology , Spinal Nerves/enzymology , Spinal Nerves/injuries , Animals , Calcium Channels, N-Type/genetics , Casein Kinase 1 epsilon/antagonists & inhibitors , Casein Kinase 1 epsilon/genetics , Disease Models, Animal , Down-Regulation/genetics , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Ganglia, Spinal/physiopathology , Hyperalgesia/enzymology , Hyperalgesia/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuralgia/enzymology , Neuralgia/physiopathology , Nociceptors/enzymology , Organ Culture Techniques , Peripheral Nervous System Diseases/physiopathology , Posterior Horn Cells/enzymology , RNA, Messenger/metabolism , Spinal Cord/physiopathology , Spinal Nerve Roots/enzymology , Spinal Nerve Roots/physiopathology , Spinal Nerves/physiopathology , Up-Regulation/physiology
7.
Genes Cells ; 14(11): 1253-69, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19817876

ABSTRACT

Spinocerebellar ataxia type 6 (SCA6) is caused by a small expansion of polyglutamine (polyQ)-encoding CAG repeat in Ca(v)2.1 calcium channel gene. To gain insights into pathogenic mechanism of SCA6, we used HEK293 cells expressing fusion protein of enhanced green fluorescent protein and Ca(v)2.1 carboxyl terminal fragment (EGFP-Ca(v)2.1CT) [L24 and S13 cells containing 24 polyQ (disease range) and 13 polyQ (normal range), respectively] and examined their responses to some stressors. When exposed to CdCl(2), L24 cells showed lower viability than the control S13 cells and caspase-dependent apoptosis was enhanced more in L24 cells. Localization of EGFP-Ca(v)2.1CT was almost confined to the nucleus, where it existed as speckle-like structures. Interestingly, CdCl(2) treatment resulted in disruption of more promyelocytic leukemia nuclear bodies (PML-NBs) in L24 cells than in S13 cells and in cells where PML-NBs were disrupted, aggregates of EGFP-Ca(v)2.1CT became larger. Furthermore, a large number of aggregates were formed in L24 cells than in S13 cells. Results of RNAi experiments indicated that HSPA1A determined the difference against CdCl(2) toxicity. Furthermore, protein expression of heat shock transcription factor 1 (HSF1), which activates HSPA1A expression, was down-regulated in L24 cells. Therefore, HSF1-HSPA1A axis is critical for the vulnerability in L24 cells.


Subject(s)
Apoptosis/physiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Models, Biological , Spinocerebellar Ataxias , Transcription Factors/genetics , Transcription Factors/metabolism , Apoptosis/drug effects , Cadmium Chloride/toxicity , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Down-Regulation , Gene Expression Regulation , Heat Shock Transcription Factors , Hot Temperature , Humans , Immunohistochemistry , Microarray Analysis , Mutation/genetics , Reverse Transcriptase Polymerase Chain Reaction , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/metabolism
8.
J Pharmacol Sci ; 110(1): 55-63, 2009 May.
Article in English | MEDLINE | ID: mdl-19403993

ABSTRACT

cDNA microarray analysis showed the expression of peripheral-type benzodiazepine receptor (PBR) mRNA is slightly enhanced in the spinal cord of mice with spinal nerve injury (SNL) as compared with sham-operated mice. PBR transports cholesterol to the mitochondria, where cholesterol is converted to pregnenolone. Pregnenolone is then metabolized to progesterone, an activator of progesterone receptor, and further metabolized to produce allopregnanolone and 3alpha,21-dihydroxy-5alpha-pregnan-20-one (3alpha,5alpha-THDOC), positive allosteric modulators and activators of the GABA(A) receptor. In the present study, we first tested whether the enhanced PBR expression is causally related to neuropathic pain, and we found that the PBR antagonist PK11195 is effective in reducing SNL-induced mechanical allodynia and thermal hyperalgesia. Next we tested whether the PK11195-induced antinociception is attributable to reduced neurosteroid synthesis, which may possibly lead to reduced activation of the progesterone receptor and/or GABA(A) receptor. We found that allopregnanolone and 3alpha,5alpha-THDOC are effective in reducing the anti-hyperalgesic effect of PK11195, suggesting a partial contribution of reduced GABA(A)-receptor activation to PK11195-induced antinociception.


Subject(s)
GABA-A Receptor Antagonists , Pain/drug therapy , Pain/etiology , Peripheral Nervous System Diseases/complications , Peripheral Nervous System Diseases/drug therapy , Analgesics/pharmacology , Animals , Behavior, Animal/drug effects , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Desoxycorticosterone/analogs & derivatives , Desoxycorticosterone/pharmacology , Immunohistochemistry , Injections, Spinal , Isoquinolines/pharmacology , Male , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Pregnanolone/pharmacology , Progesterone/pharmacology , Receptors, GABA-A/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord/metabolism , Spinal Nerves/injuries , Spinal Nerves/pathology
9.
FEBS J ; 276(5): 1221-32, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19175674

ABSTRACT

Insulin-like growth factors (IGFs) play essential roles in fetal and postnatal growth and development of mammals. They are secreted by a wide variety of tissues, with the liver being the major source of circulating IGFs, and regulate cell growth, differentiation and survival. IGFs share some biological activities with insulin but are secreted in distinct physiological and developmental contexts, having specific functions. Although recent analyses of invertebrate genomes have revealed the presence of multiple insulin family peptide genes in each genome, little is known about functional diversification of the gene products. Here we show that a novel insulin family peptide of the silkmoth Bombyx mori, which was purified and sequenced from the hemolymph, is more like IGFs than like insulin, in contrast to bombyxins, which are previously identified insulin-like peptides in B. mori. Expression analysis reveals that this IGF-like peptide is predominantly produced by the fat body, a functional equivalent of the vertebrate liver and adipocytes, and is massively released during pupa-adult development. Studies using in vitro tissue culture systems show that secretion of the peptide is stimulated by ecdysteroid and that the secreted peptide promotes the growth of adult-specific tissues. These observations suggest that this peptide is a Bombyx counterpart of vertebrate IGFs and that functionally IGF-like peptides may be more ubiquitous in the animal kingdom than previously thought. Our results also suggest that the known effects of ecdysteroid on insect adult development may be in part mediated by IGF-like peptides.


Subject(s)
Bombyx/growth & development , Ecdysteroids/pharmacology , Insect Proteins/chemistry , Somatomedins/chemistry , Amino Acid Sequence , Animals , Bombyx/metabolism , Female , Insect Proteins/genetics , Insect Proteins/isolation & purification , Intercellular Signaling Peptides and Proteins/chemistry , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/isolation & purification , Molecular Sequence Data , Peptides/chemistry , Peptides/isolation & purification , Phylogeny , Somatomedins/genetics , Somatomedins/isolation & purification
10.
Mol Cell Neurosci ; 34(2): 261-70, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17188510

ABSTRACT

Spinocerebellar ataxia type 6 (SCA6) is caused by polyglutamine expansion in P/Q-type Ca2+ channels (Ca(v)2.1) and is characterized by predominant degeneration of cerebellar Purkinje cells. To characterize the Ca(v)2.1 channel with an SCA6 mutation in cerebellar Purkinje cells, we have generated knock-in mouse models that express human Ca(v)2.1 with 28 polyglutamine repeats (disease range) and with 13 polyglutamine repeats (normal range). Patch-clamp recordings of the Purkinje cells from homozygous control or SCA6 knock-in mice revealed a non-inactivating current that is highly sensitive to a spider toxin omega-Agatoxin IVA, indicating that the human Ca(v)2.1 expressed in Purkinje cells exhibits typical P-type properties in contrast to the previous data showing Q-type properties, when it was expressed in cultured cell lines. Furthermore, the voltage dependence of activation and inactivation and current density were not different between SCA6 and control, though these properties were altered in previous reports using non-neuronal cells as expression systems. Therefore, our results do not support the notion that the alteration of the channel properties may underlie the pathogenic mechanism of SCA6.


Subject(s)
Calcium Channels, N-Type/physiology , Calcium Channels/metabolism , Gene Expression Regulation/radiation effects , Mutation/physiology , Age Factors , Animals , Behavior, Animal/physiology , Calcium Channel Blockers/pharmacology , Cells, Cultured , Cerebellum/cytology , Electric Stimulation/methods , Gene Expression Regulation/physiology , Humans , In Situ Hybridization , Membrane Potentials/genetics , Mice , Mice, Transgenic , Motor Activity/genetics , Patch-Clamp Techniques/methods , Peptides/genetics , Purkinje Cells , Reverse Transcriptase Polymerase Chain Reaction , omega-Agatoxin IVA/pharmacology
11.
Eur J Pharmacol ; 541(1-2): 44-8, 2006 Jul 10.
Article in English | MEDLINE | ID: mdl-16759651

ABSTRACT

Injury to the spinal nerves of mice induces allodynia and hyperalgesia. Intrathecal injection of the progesterone/estrogen receptor antagonist ICI 182,780 produced antinociceptive effects. Co-administration of estrogen did not reduce but tended to enhance the antinociceptive effect of ICI 182,780. On the other hand, co-administration of progesterone dose-dependently reduced the antinociceptive effect of ICI 182,780, indicating that the antinociceptive effect is through antiprogesterone receptor activity of ICI 182,780. These results suggest that spinal progesterone receptors play an important role in neuropathic pain, and that controlling the activity of progesterone receptors may be of great importance in the treatment of neuropathic pain.


Subject(s)
Estradiol/analogs & derivatives , Estrogen Antagonists/pharmacology , Neuralgia/prevention & control , Receptors, Progesterone/antagonists & inhibitors , Analgesics/administration & dosage , Analgesics/pharmacology , Animals , Behavior, Animal/drug effects , Dose-Response Relationship, Drug , Estradiol/administration & dosage , Estradiol/pharmacology , Estrogen Antagonists/administration & dosage , Estrogens/administration & dosage , Estrogens/pharmacology , Fulvestrant , Hot Temperature/adverse effects , Hyperalgesia/etiology , Hyperalgesia/physiopathology , Hyperalgesia/prevention & control , Injections, Spinal , Male , Mice , Mice, Inbred C57BL , Neuralgia/physiopathology , Pain/etiology , Pain/physiopathology , Pain/prevention & control , Pain Measurement/methods , Progesterone/administration & dosage , Progesterone/pharmacology , Receptors, Progesterone/physiology , Spinal Nerves/injuries , Spinal Nerves/physiopathology
12.
Biochem Biophys Res Commun ; 344(3): 920-5, 2006 Jun 09.
Article in English | MEDLINE | ID: mdl-16631598

ABSTRACT

Voltage-dependent Ca(2+) channels play important roles in cerebellar functions including motor coordination and learning. Since abundant expression of Ca(V)2.3 Ca(2+) channel gene in the cerebellum was detected, we searched for possible deficits in the cerebellar functions in the Ca(V)2.3 mutant mice. Behavioral analysis detected in delayed motor learning in rotarod tests in mice heterozygous and homozygous for the Ca(V)2.3 gene disruption (Ca(V)2.3+/- and Ca(V)2.3-/-, respectively). Electrophysiological analysis of mutant mice revealed perplexing results: deficit in long-term depression (LTD) at the parallel fiber Purkinje cell synapse in Ca(V)2.3+/- mice but apparently normal LTD in Ca(V)2.3-/- mice. On the other hand, the number of spikes evoked by current injection in Purkinje cells under the current-clamp mode decreased in Ca(V)2.3 mutant mice in a gene dosage-dependent manner, suggesting that Ca(V)2.3 channel contributed to spike generation in Purkinje cells. Thus, Ca(V)2.3 channel seems to play some roles in cerebellar functions.


Subject(s)
Action Potentials/physiology , Calcium Channels/metabolism , Cation Transport Proteins/metabolism , Cerebellum/physiology , Long-Term Synaptic Depression/physiology , Purkinje Cells/physiology , Animals , Calcium Channels/deficiency , Calcium Channels/genetics , Calcium Channels, R-Type , Cation Transport Proteins/deficiency , Cation Transport Proteins/genetics , Cells, Cultured , Ion Channel Gating/physiology , Mice , Mice, Knockout , Rotarod Performance Test
13.
Eur J Pharmacol ; 524(1-3): 80-3, 2005 Nov 07.
Article in English | MEDLINE | ID: mdl-16256102

ABSTRACT

Injury to the spinal nerves of mice induces mechanical allodynia and thermal hyperalgesia. In the injured spinal cord, the expression of glucocorticoid receptor mRNA was increased, whereas it was decreased in N-type Ca(2+)-channel-deficient mice, in which neuropathic pain is eliminated. Intrathecal and intraperitoneal injection of the glucocorticoid receptor antagonist RU486 produced antinociceptive effects, whereas intracerebroventricular injection was without effect. The more selective antagonist dexamethasone 21-mesylate suppressed both mechanical allodynia and thermal hyperalgesia. These results suggest that spinal glucocorticoid receptors play an important role in neuropathic pain, and that controlling the activity of glucocorticoid receptors may be of great importance in the treatment of neuropathic pain.


Subject(s)
Hyperalgesia/prevention & control , Neuralgia/prevention & control , Pain/prevention & control , Receptors, Glucocorticoid/antagonists & inhibitors , Animals , Behavior, Animal/drug effects , Calcium Channels, N-Type/deficiency , Calcium Channels, N-Type/genetics , Calcium Channels, N-Type/physiology , Dexamethasone/analogs & derivatives , Dexamethasone/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Expression Profiling , Hormone Antagonists/administration & dosage , Hormone Antagonists/pharmacology , Hyperalgesia/etiology , Injections, Intraventricular , Injections, Spinal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mifepristone/administration & dosage , Mifepristone/pharmacology , Motor Activity/drug effects , Neuralgia/etiology , Oligonucleotide Array Sequence Analysis , Pain/etiology , Pain Threshold/drug effects , Receptors, Glucocorticoid/physiology , Spinal Cord Injuries/complications , Spinal Cord Injuries/genetics , Time Factors
14.
Genes Cells ; 10(2): 87-96, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15676020

ABSTRACT

A recent report on the mechanism of synaptic targeting of Ca(v)2.2 channel suggested that this process depends upon the presence of long C-terminal tail and that protein interactions mediated by SH3-binding and PDZ-binding motifs in the tail region are important. To examine the possibility that C-terminal tail of the Ca(v)2.1 channel and the polyglutamine stretch therein are also involved in the mechanism for channel localization, we constructed several expression plasmids for human Ca(v)2.1 channel tagged with enhanced green fluorescent protein (EGFP) and introduced them into mouse hippocampal neuronal culture. HC construct encodes short version of Ca(v)2.1, and HS and HL encode Ca(v)2.1 channel with a long C-terminal tail, which contains polyglutamine tract of 13 (normal range) and 28 (SCA6 disease range) repeat units, respectively. Surprisingly, transfection with HC, HS, and HL gave essentially the same results: EGFP signal was observed in cell soma, dendrites, and the axon as well. Furthermore, mutation of the PDZ-binding motif located at the C-terminus of the long version of Ca(v)2.1, by adding FLAG tag, did not affect the localization patterns of HS and HL as well. Therefore, the C-terminal region is not indispensable for the subcellular localization of Ca(v)2.1 channel, nor expansion of polyglutamine length affected the localization of the channel. Thus, it is possible that the localization mechanism of Ca(v)2.1 channel is different from that of Ca(v)2.2, though these channels share various structural and functional characteristics.


Subject(s)
Calcium Channels, N-Type/metabolism , Neurons/metabolism , Amino Acid Sequence , Animals , Axons/metabolism , Calcium Channels, N-Type/genetics , Cells, Cultured , Chlorocebus aethiops , Dendrites/metabolism , Glutamine/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hippocampus/cytology , Humans , Mice , Molecular Sequence Data , Mutation , Recombinant Fusion Proteins/metabolism , src Homology Domains
15.
Eur J Neurosci ; 20(12): 3516-9, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15610184

ABSTRACT

Morphine is the drug of choice to treat intractable pain, although prolonged administration often causes undesirable side-effects including analgesic tolerance. It is speculated that voltage-dependent Ca(2+) channels (VDCCs) play a key role in morphine analgesia and tolerance. To examine the subtype specificity of VDCCs in these processes, we analysed mice lacking N-type (Ca(v)2.2) or R-type (Ca(v)2.3) VDCCs. Systemic morphine administration or exposure to warm water swim-stress, known to induce endogenous opioid release, resulted in greater analgesia in Ca(v)2.3(-/-) mice than in controls. Moreover, Ca(v)2.3(-/-) mice showed resistance to morphine tolerance. In contrast, Ca(v)2.2(-/-) mice showed similar levels of analgesia and tolerance to control mice. Intracerebroventricular (i.c.v.) but not intrathecal (i.t.) administration of morphine reproduced the result of systemic morphine in Ca(v)2.3(-/-) mice. Furthermore, i.c.v. administration of an R-type channel blocker potentiated morphine analgesia in wild-type mice. Thus, the inhibition of R-type Ca(2+) current could lead to high-efficiency opioid therapy without tolerance.


Subject(s)
Analgesics, Opioid/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels, R-Type/metabolism , Morphine/pharmacology , Pain Measurement/drug effects , Animals , Calcium Channels, R-Type/genetics , Calcium Channels, R-Type/physiology , Drug Tolerance/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Pain Measurement/methods
16.
Neurosci Lett ; 350(1): 41-5, 2003 Oct 16.
Article in English | MEDLINE | ID: mdl-12962913

ABSTRACT

Volatile anesthetics are known to depress excitatory synaptic transmission. Inhibition of voltage-dependent Ca2+ channels is speculated to underlie this mechanism, which remains to be clarified in vivo. We examined the sensitivity to halothane in mice lacking the N-type Ca2+ channel, a major contributor of presynaptic neurotransmitter release. Sensitivity to halothane was significantly increased in the knockout mice compared with the wild-type littermates. Halothane also depressed field excitatory postsynaptic potentials recorded from the Schaffer collateral-CA1 hippocampal synapses more greatly in the knockout mice. We further examined sleep time induced by injection of propofol, an intravenous anesthetic that mainly affects inhibitory synaptic transmission. In contrast, sensitivity to propofol was significantly decreased in the knockout mice. We suggest that inhibition of the N-type Ca2+ channel underlies mechanisms of halothane anesthesia but counteracts propofol anesthesia.


Subject(s)
Anesthetics, Inhalation/pharmacology , Anesthetics, Intravenous/pharmacology , Calcium Channels, N-Type/physiology , Halothane/pharmacology , Propofol/pharmacology , Animals , Blood Pressure/drug effects , Calcium Channels, N-Type/genetics , Dose-Response Relationship, Drug , Excitatory Postsynaptic Potentials/drug effects , Heart Rate/drug effects , Heterozygote , Hippocampus/cytology , Hippocampus/drug effects , Homozygote , In Vitro Techniques , Mice , Mice, Knockout , Pain Measurement/drug effects , Reflex/drug effects , Reflex/genetics , Sleep/drug effects
17.
Anesth Analg ; 97(1): 96-103, table of contents, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12818950

ABSTRACT

UNLABELLED: Because inhibition of voltage-dependent Ca(2+) channels can be a mechanism underlying general anesthesia, we examined sensitivities to propofol and halothane in mice lacking the R-type (Ca(v)2.3) channel widely expressed in neurons. Sleep time after propofol injection (26 mg/kg IV) and halothane MAC(RR) and MAC (50% effective concentrations for the loss of the righting reflex and for the tail pinch/withdrawal response, respectively) were determined. Significantly shorter propofol-induced sleep time (291.6 +/- 16.8 s versus 344.4 +/- 12.1 s) and larger halothane MAC(RR) (1.11% +/- 0.04% versus 0.98% +/- 0.03%) were observed in Ca(v)2.3 channel knockouts (Ca(v)2.3(-/-)) than in wild-type (Ca(v)2.3(+/+)) litter mates. To investigate the basis of the decreased anesthetic sensitivities in vivo, field excitatory postsynaptic potentials and population spikes (PSs) were recorded from Schaffer collateral CA1 synapses in hippocampal slices. Propofol (10-30 micro M) inhibited PSs by potentiating gamma-aminobutyric acid-ergic inhibition, and this potentiation was markedly smaller at 30 micro M in Ca(v)2.3(-/-) mice, possibly accounting for the decreased propofol sensitivity in vivo. Halothane (1.4%-2.2%) inhibited field excitatory postsynaptic potentials similarly in both genotypes, whereas 1%-2% halothane depressed PSs more in Ca(v)2.3(-/-) mice, suggesting the postsynaptic role of the R-type channel in the propagation of excitation and other mechanisms underlying the increased halothane MAC(RR) in Ca(v)2.3(-/-) mice. IMPLICATIONS: Because inhibition of neuronal Ca(2+) currents can be a mechanism underlying general anesthesia, we examined anesthetic sensitivities in mice lacking the R-type (Ca(v)2.3) Ca(2+) channels both in vivo and in hippocampal slices. Decreased sensitivities in mutant mice imply a possibility that agents blocking this channel may increase the requirements of anesthetics/hypnotics.


Subject(s)
Anesthetics, Inhalation/pharmacology , Anesthetics, Intravenous/pharmacology , Calcium Channels, R-Type/genetics , Calcium Channels, R-Type/physiology , Halothane/pharmacology , Propofol/pharmacology , Animals , Blood Gas Analysis , Blood Pressure/drug effects , Body Temperature/drug effects , Dose-Response Relationship, Drug , Excitatory Postsynaptic Potentials/drug effects , Heart Rate/drug effects , Hippocampus/drug effects , In Vitro Techniques , Injections, Intravenous , Mice , Mice, Knockout , Pyramidal Cells/drug effects , Sleep/drug effects , Time Factors
18.
Biochem Biophys Res Commun ; 299(2): 299-304, 2002 Nov 29.
Article in English | MEDLINE | ID: mdl-12437986

ABSTRACT

Much evidence indicates that calcium channel plays a role in cocaine-induced behavioral responses. We assessed the contributions of Ca(v)2.3 (alpha(1E)) calcium channel to cocaine effects using Ca(v)2.3 knockout mice (Ca(v)2.3-/-). Acute administration of cocaine enhanced the locomotor activity in wild-type mice (Ca(v)2.3+/+), but failed to produce any response in Ca(v)2.3-/- mice. Repeated exposure to cocaine induced the behavioral sensitization and conditioned place preference in both genotypes. Pretreatment with a D1-receptor antagonist, SCH23390, blocked the cocaine-induced place preference in Ca(v)2.3+/+ mice; however, it had no significant effect in Ca(v)2.3-/- mice. Microdialysis and RT-PCR analysis revealed that the levels of extracellular dopamine and dopamine D1 and D2 receptor mRNAs were not altered in Ca(v)2.3-/- mice. These data indicate that Ca(v)2.3 channel contributes to the locomotor-stimulating effect of cocaine, and the deletion of Ca(v)2.3 channel reveals the presence of a novel pathway leading to cocaine rewarding which is insensitive to D1 receptor antagonist.


Subject(s)
Behavior, Animal/drug effects , Calcium Channels/physiology , Cation Transport Proteins , Cocaine/pharmacology , Motor Activity/drug effects , Animals , Benzazepines/pharmacology , Calcium Channels/genetics , Calcium Channels, R-Type , Conditioning, Psychological , Dopamine/metabolism , Dopamine Antagonists/pharmacology , Kinetics , Mice , Mice, Knockout , Neostriatum/drug effects , Neostriatum/metabolism , RNA, Messenger/biosynthesis , Receptors, Dopamine D1/antagonists & inhibitors , Receptors, Dopamine D1/biosynthesis , Receptors, Dopamine D1/genetics , Receptors, Dopamine D2/biosynthesis , Receptors, Dopamine D2/genetics
19.
Neurosci Res ; 43(1): 1-7, 2002 May.
Article in English | MEDLINE | ID: mdl-12074836

ABSTRACT

Recently several mutant mouse lines lacking neuronal voltage-dependent Ca(2+) channels (VDCCs) have been established by the use of gene targeting in embryonic stem cells. Pain-related behaviors in Ca(v)2.2 (alpha(1B)) and Ca(v)2.3 (alpha(1E)) knockout mice were studied to gain further insight into the mechanism of pain transmission, where VDCCs are thought to play important roles. We review here the data from these recent studies. Ca(v)2.3-/- mice showed normal responses to acute painful stimuli, and reduced responses to the somatic inflammatory pain stimuli. Ca(v)2.3+/- mice exhibited reduced symptoms of visceral inflammatory pain. Ca(v)2.3-/- mice showed abnormal behavior related to the descending antinociceptive mechanism activated by the intraperitoneal injection of acetic acid. Ca(v)2.2-/- mice showed variable acute nociceptive responses depending on the mutant lines. However, all the lines of Ca(v)2.2-/- mice exhibited reduced responses in the phase 2 of the formalin test, suggesting a suppression of inflammatory pain. Furthermore Ca(v)2.2-/- mice showed markedly reduced neuropathic pain symptoms after spinal nerve ligation. Impaired antinociception, similar to that seen in the Ca(v)2.3-/- mice, was also observed in the Ca(v)2.2-/- mice. Therefore, it is suggested that these mutant mice could provide novel models to delineate the nociceptive and antinociceptive mechanisms.


Subject(s)
Calcium Channels, N-Type/deficiency , Calcium Channels, R-Type/deficiency , Central Nervous System/metabolism , Neural Pathways/metabolism , Neurons/metabolism , Nociceptors/metabolism , Pain/metabolism , Synaptic Transmission/physiology , Acetic Acid/pharmacology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Calcium Channels, N-Type/genetics , Calcium Channels, R-Type/genetics , Central Nervous System/physiopathology , Disease Models, Animal , Female , Hot Temperature/adverse effects , Inflammation/genetics , Inflammation/metabolism , Inflammation/physiopathology , Male , Mice , Mice, Knockout , Models, Neurological , Neural Pathways/physiopathology , Nociceptors/drug effects , Pain/genetics , Pain/physiopathology , Pain Measurement , Phenotype , Physical Stimulation
20.
FEBS Lett ; 516(1-3): 229-33, 2002 Apr 10.
Article in English | MEDLINE | ID: mdl-11959138

ABSTRACT

To know the function of the Ca2+ channel containing alpha(1)2.3 (alpha1E) subunit (Ca(v)2.3 channel) in spermatozoa, we analyzed Ca2+ transients and sperm motility using a mouse strain lacking Ca(v)2.3 channel. The averaged rising rates of Ca2+ transients induced by alpha-D-mannose-bovine serum albumin in the head region of Ca(v)2.3-/- sperm were significantly lower than those of Ca(v)2.3+/+ sperm. A computer-assisted sperm motility assay revealed that straight-line velocity and linearity were greater in Ca(v)2.3-/- sperm than those in Ca(v)2.3+/+ sperm. These results suggest that the Ca(v)2.3 channel plays some roles in Ca2+ transients and the control of flagellar movement.


Subject(s)
Calcium Channels/physiology , Cation Transport Proteins , Sperm Motility/physiology , Animals , Calcium Channels/genetics , Calcium Channels, R-Type , Calcium Signaling/genetics , Calcium Signaling/physiology , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Movement/physiology , Sperm Motility/genetics , Sperm Tail/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...