Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
Proc Natl Acad Sci U S A ; 109(33): 13368-73, 2012 Aug 14.
Article in English | MEDLINE | ID: mdl-22847418

ABSTRACT

The Na-Cl cotransporter (NCC), which is the target of inhibition by thiazides, is located in close proximity to the chloride-absorbing transporter pendrin in the kidney distal nephron. Single deletion of pendrin or NCC does not cause salt wasting or excessive diuresis under basal conditions, raising the possibility that these transporters are predominantly active during salt depletion or in response to excess aldosterone. We hypothesized that pendrin and NCC compensate for loss of function of the other under basal conditions, thereby masking the role that each plays in salt absorption. To test our hypothesis, we generated pendrin/NCC double knockout (KO) mice by crossing pendrin KO mice with NCC KO mice. Pendrin/NCC double KO mice displayed severe salt wasting and sharp increase in urine output under basal conditions. As a result, animals developed profound volume depletion, renal failure, and metabolic alkalosis without hypokalemia, which were all corrected with salt replacement. We propose that the combined inhibition of pendrin and NCC can provide a strong diuretic regimen without causing hypokalemia for patients with fluid overload, including patients with congestive heart failure, nephrotic syndrome, diuretic resistance, or generalized edema.


Subject(s)
Anion Transport Proteins/deficiency , Diuresis/physiology , Receptors, Drug/deficiency , Renal Insufficiency/physiopathology , Sodium Chloride/metabolism , Symporters/deficiency , Alkalosis/blood , Alkalosis/complications , Alkalosis/physiopathology , Animals , Anion Transport Proteins/metabolism , Body Weight , Kidney/pathology , Kidney/physiopathology , Kidney Function Tests , Mice , Mice, Knockout , Models, Biological , Potassium/blood , Receptors, Drug/metabolism , Renal Insufficiency/blood , Renal Insufficiency/complications , Sodium/blood , Sodium Chloride, Dietary , Solute Carrier Family 12, Member 3 , Sulfate Transporters , Symporters/metabolism , Vasopressins/administration & dosage , Vasopressins/pharmacology , Water-Electrolyte Balance/physiology
2.
Kidney Int ; 83(5): 811-24, 2013 May.
Article in English | MEDLINE | ID: mdl-23447069

ABSTRACT

A dietary potassium load induces a rapid kaliuresis and natriuresis, which may occur even before plasma potassium and aldosterone (aldo) levels increase. Here we sought to gain insight into underlying molecular mechanisms contributing to this response. After gastric gavage of 2% potassium, the plasma potassium concentrations rose rapidly (0.25 h), followed by a significant rise of plasma aldo (0.5 h) in mice. Enhanced urinary potassium and sodium excretion was detectable as early as spot urines could be collected (about 0.5 h). The functional changes were accompanied by a rapid and sustained (0.25-6 h) dephosphorylation of the NaCl cotransporter (NCC) and a late (6 h) upregulation of proteolytically activated epithelial sodium channels. The rapid effects on NCC were independent from the coadministered anion. NCC dephosphorylation was also aldo-independent, as indicated by experiments in aldo-deficient mice. The observed urinary sodium loss relates to NCC, as it was markedly diminished in NCC-deficient mice. Thus, downregulation of NCC likely explains the natriuretic effect of an acute oral potassium load in mice. This may improve renal potassium excretion by increasing the amount of intraluminal sodium that can be exchanged against potassium in the aldo-sensitive distal nephron.


Subject(s)
Kidney/metabolism , Potassium, Dietary/blood , Receptors, Drug/metabolism , Symporters/metabolism , Administration, Oral , Aldosterone/blood , Animals , Biological Transport , Cytochrome P-450 CYP11B2/deficiency , Cytochrome P-450 CYP11B2/genetics , Epithelial Sodium Channels/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Natriuresis , Phosphorylation , Potassium, Dietary/administration & dosage , Potassium, Dietary/urine , Receptors, Drug/deficiency , Receptors, Drug/genetics , Solute Carrier Family 12, Member 3 , Symporters/deficiency , Symporters/genetics , Time Factors , Water-Electrolyte Balance
3.
Cell Tissue Res ; 340(2): 335-46, 2010 May.
Article in English | MEDLINE | ID: mdl-20383647

ABSTRACT

Trafficking of pancreatic K(ATP) channels to the plasma membrane critically depends on masking the endoplasmic reticulum (ER) retention signals of the SUR1 and Kir6.2 subunits upon their proper assembly into functional hetero-octamers. When expressed in the absence of the partner protein, each subunit might accumulate in the ER and trigger beta-cell ER stress and oxidative stress. To test this hypothesis, Kir6.2 localisation, ER ultra-structure and ER-stress- and oxidative-stress-response gene mRNA levels were evaluated in pancreatic endocrine cells from adult wild-type (WT) and Sur1 knockout (Sur1 ( -/- )) mice. As previously reported, Kir6.2 was mainly expressed on secretory granules and at the plasma membrane of WT islet cells. In contrast, like the ER chaperone calreticulin, Kir6.2 was primarily localised in the rough endoplasmic reticulum (RER) of Sur1 ( -/- ) islet cells. ER retention of Kir6.2 was demonstrated (electron microscopy) by a significant increase in the length and Kir6.2 density of RER in Sur1 ( -/- ) vs WT islet cells. Despite Kir6.2 retention in RER, Xbp1 mRNA splicing and mRNA levels of preproinsulin and ER-stress-response genes Bip, Edem and Gadd153 were similar in WT and Sur1 ( -/- ) islets. However, mRNA levels of the antioxidant enzymes Sod1, Sod2, Gpx2 and catalase were significantly up-regulated in Sur1 ( -/- ) islets. Sequestration of Kir6.2 in RER of Sur1 ( -/- ) islet cells is thus associated with an increase in RER length and mild oxidative stress without activation of the classical ER stress response.


Subject(s)
Endoplasmic Reticulum/metabolism , Islets of Langerhans/metabolism , Potassium Channels, Inwardly Rectifying/deficiency , Potassium Channels, Inwardly Rectifying/metabolism , Receptors, Drug/deficiency , Stress, Physiological , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Antibody Specificity/immunology , Calreticulin/metabolism , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/ultrastructure , Gene Expression Regulation , Islets of Langerhans/cytology , Islets of Langerhans/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress/genetics , Potassium Channels, Inwardly Rectifying/genetics , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Drug/genetics , Receptors, Drug/metabolism , Subcellular Fractions/metabolism , Sulfonylurea Receptors
4.
Cell Tissue Res ; 335(3): 505-15, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19142666

ABSTRACT

In congenital hyperinsulinism of infancy (CHI), the loss of K-ATP channels (composed of Kir6.2 and SUR1 subunits) in beta cells induces permanent insulin secretion and severe hypoglycaemia. By contrast, Sur1 ( -/- ) mice do not present such defects. We have investigated the impact of Sur1 gene inactivation on mouse islet cell morphology, structure and basic physiology. Pancreata were collected from young, adult and old wild-type (WT) and Sur1 ( -/- ) mice. After immunostaining for hormone, the total endocrine tissue, cell proportion, cell size and intra-insular distribution, hormone content and Glut-2 expression were quantified by morphometry. Basic physiological parameters were also measured. In young Sur1 ( -/- ) mice, the total endocrine tissue and proportion of beta cells were higher (P<0.05) than in WT mice, whereas the proportion of delta cells was lower (P<0.01). In old Sur1 ( -/- ) mice, alpha cells were frequently located in the central regions of islets (unlike WT islets) and their proportion was increased (P<0.05). Glut-2 protein and mRNA levels were lower in old Sur1 ( -/- ) islets (P<0.02). Insulinaemia, fasting insulin and glucagon contents were equivalent in both groups of pancreata. Thus, the islets of Sur1 ( -/- ) mice present morphological modifications that have not been described in CHI and that might reflect an adaptive mechanism controlling insulin secretion in these mice.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Islets of Langerhans/physiology , Pancreas/physiology , Potassium Channels, Inwardly Rectifying/genetics , Receptors, Drug/genetics , Age Factors , Animals , Blood Glucose/metabolism , Female , Gene Silencing , Glucagon/genetics , Glucagon/metabolism , Immunohistochemistry , Insulin/blood , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Pancreas/anatomy & histology , Pancreas/cytology , Pancreas/metabolism , Potassium Channels, Inwardly Rectifying/deficiency , Receptors, Drug/deficiency , Sulfonylurea Receptors
5.
Neuron ; 31(3): 453-62, 2001 Aug 16.
Article in English | MEDLINE | ID: mdl-11516401

ABSTRACT

Endocannabinoids are retrograde messengers released by neurons to modulate the strength of their synaptic inputs. Endocannabinoids are thought to mediate the suppression of GABA release that follows depolarization of a hippocampal CA1 pyramidal neuron-termed "depolarization-induced suppression of inhibition" (DSI). Here, we report that DSI is absent in mice which lack cannabinoid receptor-1 (CB1). Pharmacological and kinetic evidence suggests that CB1 activation inhibits presynaptic Ca2+ channels through direct G protein inhibition. Paired recordings show that endocannabinoids selectively inhibit a subclass of synapses distinguished by their fast kinetics and large unitary conductance. Furthermore, cannabinoid-sensitive inputs are unusual among central nervous system synapses in that they use N- but not P/Q-type Ca2+ channels for neurotransmitter release. These results indicate that endocannabinoids are highly selective, rapid modulators of hippocampal inhibition.


Subject(s)
Cannabinoids/metabolism , Hippocampus/physiology , Presynaptic Terminals/physiology , Pyramidal Cells/physiology , Receptors, Drug/physiology , Receptors, Presynaptic/physiology , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels/physiology , Cannabinoid Receptor Modulators , Colforsin/pharmacology , Endocannabinoids , Female , GTP-Binding Proteins/metabolism , In Vitro Techniques , Interneurons/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Patch-Clamp Techniques , Presynaptic Terminals/drug effects , Pyramidal Cells/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Cannabinoid , Receptors, Drug/deficiency , Receptors, Drug/genetics , Receptors, Presynaptic/drug effects , Signal Transduction
6.
Neuron ; 36(1): 57-68, 2002 Sep 26.
Article in English | MEDLINE | ID: mdl-12367506

ABSTRACT

Peripheral inflammation induces p38 MAPK activation in the soma of C fiber nociceptors in the dorsal root ganglion (DRG) after 24 hr. Inflammation also increases protein, but not mRNA levels, of the heat-gated ion channel TRPV1 (VR1) in these cells, which is then transported to peripheral but not central C fiber terminals. Inhibiting p38 activation in the DRG reduces the increase in TRPV1 in the DRG and inflamed skin and diminishes inflammation-induced heat hypersensitivity without affecting inflammatory swelling or basal pain sensitivity. p38 activation in the DRG is secondary to peripheral production of NGF during inflammation and is required for NGF-induced increases in TRPV1. The activation of p38 in the DRG following retrograde NGF transport, by increasing TRPV1 levels in nociceptor peripheral terminals in a transcription-independent fashion, contributes to the maintenance of inflammatory heat hypersensitivity.


Subject(s)
Hyperalgesia/enzymology , Mitogen-Activated Protein Kinases/metabolism , Nerve Growth Factor/metabolism , Neurons, Afferent/enzymology , Receptors, Drug/deficiency , Up-Regulation/physiology , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Ganglia, Spinal/drug effects , Ganglia, Spinal/enzymology , Hyperalgesia/physiopathology , Immunohistochemistry , Inflammation/enzymology , Inflammation/physiopathology , Male , Mice , Mice, Knockout , Nerve Fibers, Unmyelinated/drug effects , Nerve Fibers, Unmyelinated/enzymology , Nerve Growth Factor/antagonists & inhibitors , Neuralgia/enzymology , Neuralgia/physiopathology , Neurons, Afferent/drug effects , Nociceptors/drug effects , Nociceptors/enzymology , Posterior Horn Cells/enzymology , Rats , Rats, Sprague-Dawley , Receptors, Drug/drug effects , Receptors, Drug/genetics , Up-Regulation/drug effects , p38 Mitogen-Activated Protein Kinases
7.
Mol Pharmacol ; 74(3): 736-43, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18511652

ABSTRACT

ATP-sensitive K+ (KATP) channel openers are vasodilators that activate both plasma membrane and mitochondrial KATP channels. Here, we investigated the molecular mechanisms by which diazoxide and pinacidil induce vasodilation by studying diameter regulation of wild-type [SUR2(+/+)] and sulfonylurea receptor (SUR) 2-deficient [SUR2(-/-)] mouse myogenic mesenteric arteries. Ryanodine (10 microM), a ryanodine-sensitive Ca2+ release (RyR) channel blocker; iberiotoxin (100 nM), a large-conductance Ca2+-activated K+ (KCa) channel blocker; 4-aminopyridine (4-AP; 1 mM), a voltage-gated K+ (KV) channel blocker; manganese(III) tetrakis(1-methyl-4-pyridyl)porphyrin (MnTMPyP; 100 microM), an antioxidant; and a combination of ryanodine and 4-AP reduced diazoxide (100 microM)-induced dilation in pressurized (60 mm Hg) SUR2(+/+) arteries by 45 to 77%. In contrast, these inhibitors did not alter pinacidil (5 microM)-induced dilation in SUR2(+/+) arteries. Reverse transcription-polymerase chain reaction indicated that SUR2B was the only SUR isoform expressed in SUR2(+/+) mesenteric artery smooth muscle cells, whereas SURs were absent in SUR2(-/-) cells. In SUR2(-/-) arteries, pinacidil-induced vasodilation was 10% of that in SUR2(+/+) arteries, whereas diazoxide-induced vasodilation was similar in SUR2(+/+) and SUR2(-/-) arteries. Atpenin (1 microM), a selective electron transport chain (ETC) complex II inhibitor, dilated arteries similarly to diazoxide, and this effect was attenuated by MnTMPyP and ryanodine + 4-AP. Atpenin also attenuated diazoxide-, but not pinacidil-induced vasodilation. In summary, data indicate that pinacidil-induced vasodilation requires SUR2B, whereas diazoxide-induced vasodilation does not require SURs. Rather, diazoxide-induced vasodilation involves ETCII inhibition; a smooth muscle cell-reactive oxygen species elevation; and RyR, KCa, and KV channel activation. These data indicate that KATP channel openers regulate arterial diameter via SUR-dependent and -independent pathways.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Adenosine Triphosphate/pharmacology , Membrane Transport Modulators/pharmacology , Potassium Channels, Inwardly Rectifying/metabolism , Potassium Channels/metabolism , Receptors, Drug/metabolism , Vasodilation/drug effects , Animals , Blood Pressure/drug effects , Diazoxide/pharmacology , Electron Transport Complex II/antagonists & inhibitors , Endothelium/drug effects , Endothelium/metabolism , In Vitro Techniques , Ion Channel Gating/drug effects , Mesenteric Arteries/cytology , Mesenteric Arteries/drug effects , Mice , Muscle Cells/drug effects , Muscle Cells/metabolism , Pinacidil/pharmacology , Potassium Channels, Inwardly Rectifying/deficiency , Protein Subunits/metabolism , Receptors, Drug/deficiency , Ryanodine Receptor Calcium Release Channel/metabolism , Sulfonylurea Receptors
8.
Nat Neurosci ; 5(5): 446-51, 2002 May.
Article in English | MEDLINE | ID: mdl-11976704

ABSTRACT

The striatum functions critically in movement control and habit formation. The development and function of cortical input to the striatum are thought to be regulated by activity-dependent plasticity of corticostriatal glutamatergic synapses. Here we show that the induction of a form of striatal synaptic plasticity, long-term depression (LTD), is dependent on activation of the CB1 cannabinoid receptor. LTD was facilitated by blocking cellular endocannabinoid uptake, and postsynaptic loading of anandamide (AEA) produced presynaptic depression. The endocannabinoid necessary for striatal LTD is thus likely to be released postsynaptically as a retrograde messenger. These findings demonstrate a new role for endocannabinoids in the induction of long-term synaptic plasticity in a circuit necessary for habit formation and motor control.


Subject(s)
Cannabinoids/metabolism , Corpus Striatum/metabolism , Neural Inhibition/physiology , Receptors, Drug/metabolism , Synapses/metabolism , Animals , Arachidonic Acids/metabolism , Arachidonic Acids/pharmacology , Cannabinoid Receptor Modulators , Cannabinoids/pharmacology , Corpus Striatum/cytology , Corpus Striatum/drug effects , Dronabinol/analogs & derivatives , Electric Stimulation , Endocannabinoids , Excitatory Postsynaptic Potentials/drug effects , In Vitro Techniques , Mice , Mice, Knockout , Neural Inhibition/drug effects , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Neurons/drug effects , Neurons/metabolism , Polyunsaturated Alkamides , Receptors, Cannabinoid , Receptors, Drug/deficiency , Receptors, Drug/genetics
9.
Nat Neurosci ; 5(9): 856-60, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12161756

ABSTRACT

In the urinary bladder, the capsaicin-gated ion channel TRPV1 is expressed both within afferent nerve terminals and within the epithelial cells that line the bladder lumen. To determine the significance of this expression pattern, we analyzed bladder function in mice lacking TRPV1. Compared with wild-type littermates, trpv1(-/-) mice had a higher frequency of low-amplitude, non-voiding bladder contractions. This alteration was accompanied by reductions in both spinal cord signaling and reflex voiding during bladder filling (under anesthesia). In vitro, stretch-evoked ATP release and membrane capacitance changes were diminished in bladders excised from trpv1(-/-) mice, as was hypoosmolality-evoked ATP release from cultured trpv1(-/-) urothelial cells. These findings indicate that TRPV1 participates in normal bladder function and is essential for normal mechanically evoked purinergic signaling by the urothelium.


Subject(s)
Adenosine Triphosphate/metabolism , Mechanoreceptors/metabolism , Neurons, Afferent/metabolism , Receptors, Drug/deficiency , Urinary Bladder/innervation , Urination/genetics , Visceral Afferents/metabolism , Acetic Acid/pharmacology , Animals , Capsaicin/pharmacology , Cells, Cultured , Immunohistochemistry , Male , Mechanoreceptors/drug effects , Mice , Mice, Knockout , Microscopy, Electron , Muscle Contraction/drug effects , Muscle Contraction/genetics , Muscle, Smooth/drug effects , Muscle, Smooth/innervation , Muscle, Smooth/physiopathology , Neurons, Afferent/drug effects , Nitric Oxide/metabolism , Physical Stimulation , Proto-Oncogene Proteins c-fos/metabolism , Receptors, Drug/drug effects , Receptors, Drug/genetics , Reflex/drug effects , Reflex/genetics , Signal Transduction/physiology , Spinal Cord/cytology , Spinal Cord/metabolism , Spinal Cord/physiopathology , Urinary Bladder/drug effects , Urinary Bladder/physiopathology , Urination/drug effects , Urothelium/innervation , Urothelium/pathology , Urothelium/ultrastructure , Visceral Afferents/drug effects
10.
J Clin Invest ; 112(3): 423-31, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12897210

ABSTRACT

The cannabinoid receptor type 1 (CB1) and its endogenous ligands, the endocannabinoids, are involved in the regulation of food intake. Here we show that the lack of CB1 in mice with a disrupted CB1 gene causes hypophagia and leanness. As compared with WT (CB1+/+) littermates, mice lacking CB1 (CB1-/-) exhibited reduced spontaneous caloric intake and, as a consequence of reduced total fat mass, decreased body weight. In young CB1-/- mice, the lean phenotype is predominantly caused by decreased caloric intake, whereas in adult CB1-/- mice, metabolic factors appear to contribute to the lean phenotype. No significant differences between genotypes were detected regarding locomotor activity, body temperature, or energy expenditure. Hypothalamic CB1 mRNA was found to be coexpressed with neuropeptides known to modulate food intake, such as corticotropin-releasing hormone (CRH), cocaine-amphetamine-regulated transcript (CART), melanin-concentrating hormone (MCH), and preproorexin, indicating a possible role for endocannabinoid receptors within central networks governing appetite. CB1-/- mice showed significantly increased CRH mRNA levels in the paraventricular nucleus and reduced CART mRNA levels in the dorsomedial and lateral hypothalamic areas. CB1 was also detected in epidydimal mouse adipocytes, and CB1-specific activation enhanced lipogenesis in primary adipocyte cultures. Our results indicate that the cannabinoid system is an essential endogenous regulator of energy homeostasis via central orexigenic as well as peripheral lipogenic mechanisms and might therefore represent a promising target to treat diseases characterized by impaired energy balance.


Subject(s)
Appetite/physiology , Cannabinoids/metabolism , Energy Metabolism , Fatty Acids, Unsaturated/physiology , Lipids/biosynthesis , Receptors, Drug/physiology , Adipocytes/metabolism , Animals , Cannabinoid Receptor Modulators , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/physiology , Eating/physiology , Gene Expression , Hypothalamus/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuropeptides/genetics , Neuropeptides/physiology , Obesity/physiopathology , Obesity/therapy , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Cannabinoid , Receptors, Drug/deficiency , Receptors, Drug/genetics , Thinness/physiopathology
11.
J Neurosci ; 24(28): 6410-5, 2004 Jul 14.
Article in English | MEDLINE | ID: mdl-15254097

ABSTRACT

Vanilloid receptor 1 (TRPV1) has been proposed to be the principal heat-responsive channel for nociceptive neurons. The skin of both rat and mouse receives major projections from primary sensory afferents that bind the plant lectin isolectin B4 (IB4). The majority of IB4-positive neurons are known to be heat-responsive nociceptors. Previous studies suggested that, unlike rat, mouse IB4-positive cutaneous afferents did not express TRPV1 immunoreactivity. Here, multiple antisera were used to confirm that mouse and rat have different distributions of TRPV1 and that TRPV1 immunoreactivity is absent in heat-sensitive nociceptors. Intracellular recording in TRPV1(-/-) mice was then used to confirm that TRPV1 was not required for detecting noxious heat. TRPV1(-/-) mice had more heat-sensitive neurons, and these neurons had normal temperature thresholds and response properties. Moreover, in TRPV1(-/-) mice, 82% of heat-responsive neurons did not express immunoreactivity for TRPV2, another putative noxious heat channel.


Subject(s)
Calcium Channels/deficiency , Ganglia, Spinal/cytology , Hot Temperature , Ion Channels/deficiency , Neurons, Afferent/chemistry , Nociceptors/physiology , Receptors, Drug/deficiency , Amino Acid Sequence , Animals , Calcium Channels/genetics , Calcium Channels/physiology , Epidermis/physiology , Female , Ion Channels/genetics , Ion Channels/physiology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Molecular Sequence Data , Neurons, Afferent/physiology , Nociceptors/chemistry , Plant Lectins/analysis , Rats , Rats, Sprague-Dawley , Receptors, Drug/genetics , Receptors, Drug/physiology , Species Specificity , TRPV Cation Channels
12.
J Neurosci ; 23(31): 10146-53, 2003 Nov 05.
Article in English | MEDLINE | ID: mdl-14602831

ABSTRACT

Cell death mechanisms frequently involve the influx of extracellular calcium through voltage- and ligand-gated ion channels, e.g., the NMDA receptor (Greene, 1999). The vanilloid receptor (VR1) is present in regions of the brain (Mezey et al., 2000) that are highly susceptible to neurodegenerative insults, suggesting that this ion channel might contribute to the cellular processes involved in neuronal death. We tested the effects of VR1 ligands in the oxygen glucose deprivation (OGD) model of cell death in organotypic hippocampal slice cultures. The VR1 agonist capsaicin at concentrations that are selective for VR1 did not affect cell viability per se or the extent of neurodegeneration induced by the OGD insult. In contrast, the VR1 antagonist capsazepine (0.1-10 microm) significantly reduced the amount of OGD-induced cell death. However, capsazepine was still neuroprotective in slices prepared from VR1 knock-out mice, which exhibited the same degree of neurodegeneration to that observed in slices prepared from wild-type mice, excluding the possibility that it afforded neuroprotection through inhibition of VR1. Instead, capsazepine inhibited the hyperpolarization-activated nonspecific cation channel generated current I(h) in a concentration range similar to that which was neuroprotective. Furthermore, the specific I(h) blocker ZD-7288 was also neuroprotective, mirroring the effects of capsazepine, in that it was effective at preventing cell death when applied either during or after the OGD insult. These results demonstrate that capsazepine affords neuroprotection through inhibition of I(h) rather than inhibition of VR1.


Subject(s)
Capsaicin/analogs & derivatives , Capsaicin/pharmacology , Cell Hypoxia/physiology , Glucose/metabolism , Ion Channels/antagonists & inhibitors , Nerve Tissue Proteins , Neurons/drug effects , Neurons/metabolism , Animals , Cell Death , Cells, Cultured , Cyclic Nucleotide-Gated Cation Channels , Cytoprotection/drug effects , Cytoprotection/physiology , Dose-Response Relationship, Drug , Glucose/deficiency , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , In Vitro Techniques , Ion Channels/metabolism , Mice , Mice, Knockout , Neuroprotective Agents/pharmacology , Patch-Clamp Techniques , Potassium Channels , Rats , Rats, Sprague-Dawley , Receptors, Drug/deficiency , Receptors, Drug/genetics
13.
J Neurosci ; 23(6): 2453-8, 2003 Mar 15.
Article in English | MEDLINE | ID: mdl-12657705

ABSTRACT

Although many people drink alcohol regularly, only some become addicted. Several studies have shown that genetic and environmental factors contribute to individual differences in the vulnerability to the effects of alcohol (Nestler, 2000; Kreek, 2001; Crabbe, 2002). Among the environmental factors, stress is perhaps the most important trigger for relapse after a period of abstinence (Koob and Nestler, 1997; Piazza and Le Moal, 1998; Koob and Le Moal, 2001; Weiss et al., 2001). Here we show that ethanol withdrawal symptoms were completely absent in cannabinoid CB1 receptor-deficient mice, although acute effects of ethanol and ethanol tolerance and preference were basically normal. Furthermore, foot-shock stress had no affect on alcohol preference in Cnr1-/- mice, although it induced a dramatic increase in Cnr1+/+ animals. These results reveal a critical role for the CB1 receptor in clinically important aspects of alcohol dependence and provide a rationale for the use of CB1 receptor antagonists in the treatment of alcohol addiction.


Subject(s)
Alcohol Drinking/genetics , Alcoholism/genetics , Ethanol/adverse effects , Receptor, Cannabinoid, CB1/deficiency , Receptors, Drug/deficiency , Substance Withdrawal Syndrome/physiopathology , Animals , Antineoplastic Combined Chemotherapy Protocols , Ataxia/chemically induced , Behavior, Animal/drug effects , Choice Behavior/drug effects , Choice Behavior/physiology , Cisplatin , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Tolerance/physiology , Electroshock , Ethanol/pharmacology , Hypothermia/chemically induced , Ifosfamide , Male , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitomycin , Motor Activity/drug effects , Receptor, Cannabinoid, CB1/genetics , Receptors, Cannabinoid , Receptors, Drug/genetics , Stress, Physiological , Vindesine
14.
J Neurosci ; 21(24): RC188, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11734603

ABSTRACT

Endocannabinoids are key intercellular signaling molecules in the brain, but the physiological regulation of the endocannabinoid system is not understood. We used the retrograde signal process called depolarization-induced suppression of inhibition (DSI) to study the regulation of this system. DSI is produced when an endocannabinoid released from pyramidal cells suppresses IPSCs by activating CB1R cannabinoid receptors located on inhibitory interneurons. We now report that activation of group I metabotropic glutamate receptors (mGluRs) enhances DSI and that this effect is blocked by antagonists of both mGluRs and of CB1R. We also found that DSI is absent in CB1R knock-out (CB1R(-/-)) mice, and, strikingly, that mGluR agonists have no effect on IPSCs in these mice. We conclude that group I mGluR-induced enhancement of DSI, and suppression of IPSCs, is actually mediated by endocannabinoids. This surprising result opens up new approaches to the investigation of cannabinoid actions in the brain.


Subject(s)
Cannabinoids/metabolism , Hippocampus/metabolism , Receptors, Metabotropic Glutamate/metabolism , Animals , Cannabinoid Receptor Modulators , Cannabinoids/antagonists & inhibitors , Dose-Response Relationship, Drug , Endocannabinoids , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Hippocampus/cytology , In Vitro Techniques , Interneurons/metabolism , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Knockout , Neural Inhibition/drug effects , Neural Inhibition/physiology , Patch-Clamp Techniques , Piperidines/pharmacology , Pyramidal Cells/metabolism , Pyrazoles/pharmacology , Rats , Receptor, Metabotropic Glutamate 5 , Receptors, Cannabinoid , Receptors, Drug/antagonists & inhibitors , Receptors, Drug/deficiency , Receptors, Drug/metabolism , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/antagonists & inhibitors
15.
J Neurosci ; 22(22): 9771-5, 2002 Nov 15.
Article in English | MEDLINE | ID: mdl-12427832

ABSTRACT

Endogenous cannabinoid signaling pathways have been implicated in protection of the brain from hypoxia, ischemia, and trauma, but the mechanism for these protective effects is uncertain. We found that in CB1 cannabinoid receptor knock-out mice, mortality from permanent focal cerebral ischemia was increased, infarct size and neurological deficits after transient focal cerebral ischemia were more severe, cerebral blood flow in the ischemic penumbra during reperfusion was reduced, and NMDA neurotoxicity was increased compared with wild-type littermates. These findings indicate that endogenous cannabinoid signaling pathways protect mice from ischemic stroke by a mechanism that involves CB1 receptors, and suggest that both blood vessels and neurons may be targets of this protective effect.


Subject(s)
Brain/blood supply , Brain/pathology , Receptors, Drug/deficiency , Stroke/pathology , Stroke/physiopathology , Animals , Blood Flow Velocity/genetics , Blood Pressure , Brain/drug effects , Cerebrovascular Circulation/genetics , Disease Models, Animal , Genotype , Heterozygote , Homozygote , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Male , Mice , Mice, Knockout , N-Methylaspartate/toxicity , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Phenotype , Receptors, Cannabinoid , Receptors, Drug/genetics , Reperfusion , Survival Rate
16.
J Neurosci ; 22(5): 1690-7, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11880498

ABSTRACT

Action potential firing or depolarization of the postsynaptic neuron can induce a transient suppression of inhibitory synaptic inputs to the depolarized neuron in the cerebellum and hippocampus. This phenomenon, termed depolarization-induced suppression of inhibition (DSI), is initiated postsynaptically by an elevation of intracellular Ca2+ concentration ([Ca2+]i) and is expressed presynaptically as a suppression of the transmitter release. It is, therefore, thought that some retrograde signal must exist from the depolarized postsynaptic neurons to the presynaptic terminals. Recent studies on hippocampal neurons have revealed that endogenous cannabinoids (endocannabinoids) play a key role as a retrograde messenger. There are, however, conflicting reports that glutamate may be a candidate retrograde messenger for cerebellar DSI that acts on presynaptic group II metabotropic glutamate receptors (mGluRs). In this study, we examined whether endocannabinoids mediate retrograde signal for cerebellar DSI. We recorded IPSCs from Purkinje cells by stimulating putative basket cell axons in mouse cerebellar slices. DSI was readily induced in evoked IPSCs by a depolarizing pulse train. We found that DSI was completely occluded by a cannabinoid agonist, WIN55,212-2, was totally eliminated by a specific antagonist of the type 1 cannabinoid (CB1) receptor, SR141716A, and was deficient in the CB1 knock-out mouse. In contrast, a group II mGluR-specific agonist, (2S,2'R,3'R)-2-(2',3'-dicarboxycyclopropyl)glycine, did not completely occlude DSI, and an mGluR antagonist, (RS)-alpha-methyl-4-carboxyphenylglycine, had no depressant effect on DSI. These results clearly indicate that the CB1 receptor mediates retrograde signal for DSI in cerebellar Purkinje cells.


Subject(s)
Cerebellum/metabolism , Neural Inhibition/physiology , Purkinje Cells/metabolism , Receptors, Drug/metabolism , Signal Transduction/physiology , Animals , Benzoxazines , Calcium/metabolism , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cannabinoid Receptor Modulators , Cannabinoids/pharmacology , Cells, Cultured , Cerebellum/cytology , Cerebellum/drug effects , Electric Stimulation , Fluorescent Dyes , In Vitro Techniques , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Morpholines/pharmacology , Naphthalenes/pharmacology , Neural Inhibition/drug effects , Patch-Clamp Techniques , Piperidines/pharmacology , Purkinje Cells/drug effects , Pyrazoles/pharmacology , Receptors, Cannabinoid , Receptors, Drug/deficiency , Receptors, Drug/drug effects , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/metabolism , Rimonabant , Signal Transduction/drug effects
17.
J Neurosci ; 22(10): 3864-72, 2002 May 15.
Article in English | MEDLINE | ID: mdl-12019305

ABSTRACT

Recent studies have clarified that endogenous cannabinoids (endocannabinoids) are released from depolarized postsynaptic neurons in a Ca(2+)-dependent manner and act retrogradely on presynaptic cannabinoid receptors to suppress inhibitory or excitatory neurotransmitter release. This type of modulation has been found in the hippocampus and cerebellum and was called depolarization-induced suppression of inhibition (DSI) or excitation (DSE). In this study, we quantitatively examined the effects of postsynaptic depolarization and a cannabinoid agonist on excitatory and inhibitory synapses in rat hippocampal slices and cultures. We found that both DSE and DSI can be induced, but DSE was much less prominent than DSI. For the induction of DSE, the necessary duration of depolarization was longer than for DSI. The magnitude of DSE was much smaller than that of DSI. To explore the reasons for these differences, we tested the sensitivity of EPSCs and IPSCs to a cannabinoid agonist, WIN55,212-2, in hippocampal cultures. IPSCs were dichotomized into two distinct populations, one with a high sensitivity to WIN55,212-2 (50% block at 2 nm) and the other with no sensitivity. In contrast, EPSCs were homogeneous and exhibited a low sensitivity to WIN55,212-2 (50% block at 60 nm). We estimated that the 5 sec depolarization elevated the local endocannabinoid concentration to a level equivalent to several nanomoles of WIN55,212-2. Using CB1 knock-out mice, we verified that both DSI and DSE were mediated by the cannabinoid CB1 receptor. These results indicate that presynaptic cannabinoid sensitivity is a major factor that determines the extent of DSI and DSE.


Subject(s)
Cannabinoids/pharmacology , Hippocampus/drug effects , Hippocampus/physiology , Synapses/drug effects , Synapses/physiology , Animals , Benzoxazines , Calcium Channel Blockers/pharmacology , Cannabinoid Receptor Modulators , Cells, Cultured , Dose-Response Relationship, Drug , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Hippocampus/cytology , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Morpholines/pharmacology , Naphthalenes/pharmacology , Neural Inhibition/drug effects , Neural Inhibition/physiology , Neurons/physiology , Patch-Clamp Techniques , Piperidines/pharmacology , Presynaptic Terminals/drug effects , Presynaptic Terminals/physiology , Pyrazoles/pharmacology , Rats , Receptors, Cannabinoid , Receptors, Drug/antagonists & inhibitors , Receptors, Drug/deficiency , Receptors, Drug/genetics , Rimonabant
18.
J Neurosci ; 23(14): 6058-62, 2003 Jul 09.
Article in English | MEDLINE | ID: mdl-12853424

ABSTRACT

The capsaicin receptor transient receptor potential V1 (TRPV1; also known as vanilloid receptor 1) is a sensory neuron-specific ion channel that serves as a polymodal detector of pain-producing chemical and physical stimuli. It has been reported that extracellular ATP potentiates the TRPV1 currents evoked by capsaicin or protons and reduces the temperature threshold for its activation through metabotropic P2Y receptors in a PKC-dependent pathway, suggesting that TRPV1 activation could trigger the sensation of pain at normal body temperature in the presence of ATP. Here, we show that ATP-induced thermal hyperalgesia was abolished in mice lacking TRPV1, suggesting the functional interaction between ATP and TRPV1 at a behavioral level. However, thermal hyperalgesia was preserved in P2Y1 receptor-deficient mice. Patch-clamp analyses using mouse dorsal root ganglion neurons indicated the involvement of P2Y2 rather than P2Y1 receptors. Coexpression of TRPV1 mRNA with P2Y2 mRNA, but not P2Y1 mRNA, was determined in the rat lumbar DRG using in situ hybridization histochemistry. These data indicate the importance of metabotropic P2Y2 receptors in nociception through TRPV1.


Subject(s)
Adenosine Triphosphate/pharmacology , Hypesthesia/genetics , Receptors, Drug/metabolism , Receptors, Purinergic P2/metabolism , Animals , Behavior, Animal/physiology , Capsaicin/pharmacology , Cells, Cultured , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Hot Temperature/adverse effects , Hypesthesia/chemically induced , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Pain Measurement , Patch-Clamp Techniques , RNA, Messenger/metabolism , Receptors, Drug/deficiency , Receptors, Drug/genetics , Receptors, Purinergic P2/deficiency , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2Y1 , Receptors, Purinergic P2Y2 , TRPV Cation Channels , Uridine Triphosphate/pharmacology
19.
J Neurosci ; 24(18): 4293-9, 2004 May 05.
Article in English | MEDLINE | ID: mdl-15128843

ABSTRACT

Proteinase-activated receptor (PAR) 2 is expressed on a subset of primary afferent neurons and involved in inflammatory nociception. Transient receptor potential vanilloid subfamily 1 (TRPV1) is a sensory neuron-specific cation channel that responds to capsaicin, protons, or heat stimulus. Here, we show that TRPV1 is coexpressed with PAR2 but not with PAR1 or PAR3, and that TRPV1 can functionally interact with PAR2. In human embryonic kidney 293 cells expressing TRPV1 and PAR2, PAR2 agonists increased capsaicin- or proton-evoked TRPV1 currents through a PKC-dependent pathway. After application of PAR2 agonists, temperature threshold for TRPV1 activation was reduced from 42 degrees C to well below the body temperature. PAR2-mediated Fos expression in spinal cord was decreased in TRPV1-deficient mice. The functional interaction was also observed in mouse DRG neurons and proved at a behavioral level. These represent a novel mechanism through which trypsin or tryptase released in response to tissue inflammation might trigger the sensation of pain by PAR2 activation.


Subject(s)
Endopeptidases/metabolism , Inflammation/physiopathology , Pain/physiopathology , Receptor, PAR-2/metabolism , Receptors, Drug/metabolism , Animals , Cells, Cultured , Enzyme Inhibitors/pharmacology , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Humans , Hyperalgesia/genetics , Hyperalgesia/physiopathology , Inflammation/enzymology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Pain/enzymology , Patch-Clamp Techniques , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Receptor, PAR-1/genetics , Receptor, PAR-1/metabolism , Receptor, PAR-2/agonists , Receptor, PAR-2/genetics , Receptors, Drug/deficiency , Receptors, Drug/genetics , Receptors, Thrombin/genetics , Receptors, Thrombin/metabolism , Signal Transduction/physiology , Spinal Cord/metabolism , Spinal Cord/pathology , Temperature , Transfection
20.
J Neurosci ; 22(23): 10182-91, 2002 Dec 01.
Article in English | MEDLINE | ID: mdl-12451119

ABSTRACT

Endogenous cannabinoids (endocannabinoids) are endogenous compounds that resemble the active ingredient of marijuana and activate the cannabinoid receptor in the brain. They mediate retrograde signaling from principal cells to both inhibitory ["depolarization-induced suppression of inhibition" (DSI)] and excitatory ("depolarization-induced suppression of excitation") afferent fibers. Transient endocannabinoid release is triggered by voltage-dependent Ca(2+) influx and is upregulated by group I metabotropic glutamate receptor activation. Here we show that muscarinic acetylcholine receptor (mAChR) activation also enhances transient endocannabinoid release (DSI) and induces persistent release. Inhibitory synapses in the rat hippocampal CA1 region of acute slices were studied using whole-cell patch-clamp techniques. We found that low concentrations (0.2-0.5 microm) of carbachol (CCh) enhanced DSI without affecting basal evoked IPSCs (eIPSCs) by activating mAChRs on postsynaptic cells. Higher concentrations of CCh (> or =1 microm) enhanced DSI and also persistently depressed basal eIPSCs, mainly by releasing endocannabinoids. Persistent CCh-induced endocannabinoid release did not require an increase in [Ca2+]i but was dependent on G-proteins. Although they were independent at the receptor level, muscarinic and glutamatergic mechanisms of endocannabinoid release shared intracellular machinery. Replication of the effects of CCh by blocking acetylcholinesterase with eserine suggests that mAChR-mediated endocannabinoid release is physiologically relevant. This study reveals a new role of the muscarinic cholinergic system in mammalian brain.


Subject(s)
Cannabinoids/metabolism , Hippocampus/metabolism , Receptors, Muscarinic/metabolism , Animals , Calcium/metabolism , Cannabinoid Receptor Modulators , Carbachol/pharmacology , Cholinergic Agonists/pharmacology , Cholinesterase Inhibitors/pharmacology , Dose-Response Relationship, Drug , Evoked Potentials/drug effects , Evoked Potentials/physiology , GTP-Binding Proteins/metabolism , Hippocampus/cytology , Hippocampus/drug effects , In Vitro Techniques , Interneurons/drug effects , Interneurons/metabolism , Male , Mice , Mice, Knockout , Neural Inhibition/drug effects , Neural Inhibition/physiology , Patch-Clamp Techniques , Piperidines/pharmacology , Pyrazoles/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Cannabinoid , Receptors, Drug/deficiency , Receptors, Drug/genetics , Receptors, Metabotropic Glutamate/metabolism , Receptors, Muscarinic/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL