Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
Add more filters










Publication year range
1.
Br J Pharmacol ; 153(1): 140-7, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18026125

ABSTRACT

BACKGROUND AND PURPOSE: Sphingosine 1-phosphate (S1P) selectively and potently constricts isolated cerebral arteries, but this response has not been pharmacologically characterized. EXPERIMENTAL APPROACH: The receptor subtype(s) involved in S1P-induced cerebrovascular constriction were characterized using genetic (S1P(2) and S1P(3) receptor null mice) and pharmacological tools (phospho-FTY720, a S1P(1/3/4/5) receptor agonist; SEW2871, a S1P(1) receptor agonist, JTE-013, a S1P(2) receptor antagonist, VPC23019, a S1P(1/3) receptor antagonist). Isolated basilar or peripheral (femoral, mesenteric resistance) arteries, from either rat or mouse, were studied in a wire myograph. KEY RESULTS: S1P concentration-dependently constricted basilar artery in rat, wild-type (WT) and S1P(2) null mice, but barely affected vascular tone in S1P(3) null mice. Vasoconstriction to U46619 (a thromboxane analogue) or to endothelin-1 did not differ between WT, S1P(2) and S1P(3) null mice. JTE-013 inhibited not only S1P-induced vasoconstriction, but also KCl-, U46619- and endothelin-1-induced constriction. This effect was observed in WT as well as in S1P(2) null mice. VPC23019 increased the concentration-dependent vasoconstriction to S1P in both rat and mouse basilar arteries with intact endothelium, but not in rat basilar artery without endothelium. Phospho-FTY720 concentration-dependently constricted rat basilar arteries, but not femoral or mesenteric resistance arteries, while SEW2871 did not induce any response in the same arteries. CONCLUSIONS AND IMPLICATIONS: S1P constricts cerebral arteries through S1P(3) receptors. The purported S1P(2) receptor antagonist JTE-013 does not appear to be selective, at least in rodents. Enhancement of S1P-induced contraction by VPC23019 might be related to blockade of S1P(1) receptors and NO generation.


Subject(s)
Cerebral Arteries/drug effects , Receptors, Lysosphingolipid/physiology , Vasoconstriction/drug effects , Animals , Cerebral Arteries/physiology , Dose-Response Relationship, Drug , Fingolimod Hydrochloride , In Vitro Techniques , Lysophospholipids/pharmacology , Male , Mice , Mice, Inbred C57BL , Oxadiazoles/pharmacology , Propylene Glycols/pharmacology , Pyrazoles/pharmacology , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Thiophenes/pharmacology
2.
Cephalalgia ; 24(5): 398-407, 2004 May.
Article in English | MEDLINE | ID: mdl-15096229

ABSTRACT

Triptans are commonly used anti-migraine drugs and show agonist action mainly at serotonin 5-HT(1B/1D/1F) receptors. It is not known whether frequent or long-term treatment with these drugs would alter 5-HT receptor function. We investigated the effects of protracted (14-18 days) sumatriptan and zolmitriptan treatment in rats on 5-HT(1) receptor mRNA expression and function in tissues related to migraine pathophysiology. RT-PCR analysis revealed that 5-HT(1B/1D/1F) receptor mRNA was reduced in the trigeminal ganglion after treatment with either triptan (reduction by: sumatriptan 39% and zolmitriptan 61% for 5-HT(1B); 60%vs 41% for 5-HT(1D); 32%vs 68% for 5-HT(1F)). Sumatriptan attenuated 5-HT(1D) receptor mRNA by 49% in the basilar artery, whereas zolmitriptan reduced 5-HT(1B) mRNA in this tissue by 70%. No change in 5-HT(1) receptor mRNA expression was observed in coronary artery and dura mater. Chronic triptan treatment had no effect in two functional assays [sumatriptan mediated inhibition (50 mg/kg, i.p.) of electrically induced plasma protein extravasation in dura mater and 5-nonyloxytryptamine-stimulated [(35)S]guanosine-5'-O-(3-thio)triphosphate binding in substantia nigra]. Furthermore, vasoconstriction to 5-HT in isolated basilar artery was not affected by chronic triptan treatment, while it was slightly reduced in coronary artery. We conclude that, although our treatment protocol altered mRNA receptor expression in several tissues relevant to migraine pathophysiology, it did not attenuate 5-HT(1) receptor-dependent functions in rats.


Subject(s)
Oxazolidinones/pharmacology , Receptors, Serotonin, 5-HT1/drug effects , Serotonin Receptor Agonists/pharmacology , Sumatriptan/pharmacology , Animals , Autoradiography , Basilar Artery/drug effects , Basilar Artery/metabolism , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Dura Mater/drug effects , Dura Mater/metabolism , Male , Organ Culture Techniques , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Trigeminal Ganglion/drug effects , Trigeminal Ganglion/metabolism , Tryptamines , Vasoconstriction/drug effects
3.
Cephalalgia ; 22(5): 384-94, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12110114

ABSTRACT

In order to investigate the c-fos response within the trigeminal nucleus caudalis (Sp5C) after noxious meningeal stimulation, capsaicin (0.25, 0.5, 1 and 5 nmol) was administered intracisternally in urethane (1 g/kg) and alpha-chloralose (20 mg/kg) anaesthetized male mice. Capsaicin induced a robust and dose-dependent c-fos-like immunoreactivity (c-fos LI) within Sp5C. C-fos LI was observed within laminae I and II of the entire brain stem from the area postrema to C2 level, being maximum at the decussatio pyramidum level. The area postrema, solitary tract, medullary and lateral reticular nuclei were also labelled. The 5-hydroxytryptamine(1B/1D/1F) receptor agonist sumatriptan (0.01, 0.1, 1 and 10 mg/kg), administered intraperitoneally 15 min before capsaicin stimulation (1 nmol), decreased the c-fos response within Sp5C, but not within solitary tract. The novel specific 5-hydroxytryptamine1F agonist LY 344864 (0.1 and 1 mg/kg, i.p.) significantly decreased the c-fos LI within the Sp5C as well. These findings suggest that intracisternally administered capsaicin activates the trigeminovascular system and that the pain neurotransmission can be modulated by 5-hydroxytryptamine(1B/1D/1F) receptors in mice. Thus, the availability of this model in mice, taken together with the possibility of altering the expression of specific genes in this species, may help to investigate further the importance of distinct proteins in the neurotransmission of cephalic pain.


Subject(s)
Capsaicin/pharmacology , Gene Expression Regulation/drug effects , Nerve Tissue Proteins/biosynthesis , Proto-Oncogene Proteins c-fos/biosynthesis , Receptors, Serotonin/drug effects , Trigeminal Caudal Nucleus/drug effects , Anesthetics, General , Animals , Area Postrema/metabolism , Brain Stem/metabolism , Capsaicin/administration & dosage , Capsaicin/antagonists & inhibitors , Carbazoles/pharmacology , Chloralose , Cisterna Magna , Dose-Response Relationship, Drug , Fluorobenzenes/pharmacology , Genes, fos , Hemodynamics/drug effects , Injections , Male , Mice , Models, Animal , Pain/physiopathology , Receptor, Serotonin, 5-HT1B , Receptor, Serotonin, 5-HT1D , Receptors, Serotonin/physiology , Serotonin Receptor Agonists/pharmacology , Sumatriptan/pharmacology , Synaptic Transmission/drug effects , Trigeminal Caudal Nucleus/metabolism , Urethane , Vasomotor System/physiopathology , Receptor, Serotonin, 5-HT1F
4.
Brain ; 124(Pt 12): 2490-502, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11701602

ABSTRACT

Nitric oxide (NO) has been implicated in migraine pathogenesis based on the delayed development of typical migraine headache 4-6 h after infusing the NO donor nitroglycerin [glyceryl trinitrate (GTN)] to migraineurs. Furthermore, inhibiting the synthesis of NO by treatment with a NO synthase (NOS) inhibitor attenuates spontaneous migraine headaches in 67% of subjects. Because NO has been linked to inflammation and cytokine expression, we investigated the delayed consequences of brief GTN infusion (30 min) on the development of meningeal inflammation in a rat model using doses relevant to the human model. We found dose-dependent Type II NOS [inducible NOS (iNOS)] mRNA upregulation in dura mater beginning at 2 h and an increase in the corresponding protein expression at 4, 6 and 10 h after infusion. Type II NOS immunoreactivity was expressed chiefly within resident meningeal macrophages. Consistent with development of a delayed inflammatory response, we detected induction of interleukin 1beta in dura mater at 2 and 6 h and increased interleukin 6 in dural macrophages and in rat cerebrospinal fluid at 6 h after GTN infusion. Myeloperoxidase-positive cells were rarely found. Leakage of plasma proteins from dural blood vessels was first detected 4 h after GTN infusion, and this was suppressed by administering a specific Type II NOS inhibitor [L-N(6)-(1-iminoethyl)-lysine (L-NIL)]. In addition to cytokine induction, macrophage iNOS upregulation and oedema formation after GTN infusion, dural mast cells exhibited granular changes consistent with secretion at 4 and 6 h. Because iNOS was expressed in dural macrophages following topical GTN, and in the spleen after intravenous injection, the data suggest that the inflammatory response is mediated by direct actions on the dura and does not develop secondary to events within the brain. Our findings point to the importance of new gene expression and cytokine expression as fundamental to the delayed response following GTN infusion, and support the hypothesis that a similar response develops in human meninges after GTN challenge.


Subject(s)
Meningitis/immunology , Meningitis/physiopathology , Migraine Disorders/immunology , Migraine Disorders/physiopathology , Animals , Blood Proteins/metabolism , Dura Mater/immunology , Dura Mater/metabolism , Gene Expression/immunology , Interleukin-1/analysis , Interleukin-1/biosynthesis , Interleukin-6/analysis , Interleukin-6/biosynthesis , Macrophages/chemistry , Macrophages/immunology , Macrophages/metabolism , Male , Mast Cells/chemistry , Mast Cells/drug effects , Mast Cells/immunology , Nitric Oxide/immunology , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Nitroglycerin/pharmacology , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Vasodilator Agents/pharmacology
5.
J Neurosci ; 21(14): 5045-53, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11438580

ABSTRACT

After mild ischemic insults, many neurons undergo delayed neuronal death. Aberrant activation of the cell cycle machinery is thought to contribute to apoptosis in various conditions including ischemia. We demonstrate that loss of endogenous cyclin-dependent kinase (Cdk) inhibitor p16(INK4a) is an early and reliable indicator of delayed neuronal death in striatal neurons after mild cerebral ischemia in vivo. Loss of p27(Kip1), another Cdk inhibitor, precedes cell death in neocortical neurons subjected to oxygen-glucose deprivation in vitro. The loss of Cdk inhibitors is followed by upregulation of cyclin D1, activation of Cdk2, and subsequent cytoskeletal disintegration. Most neurons undergo cell death before entering S-phase, albeit a small number ( approximately 1%) do progress to the S-phase before their death. Treatment with Cdk inhibitors significantly reduces cell death in vitro. These results show that alteration of cell cycle regulatory mechanisms is a prelude to delayed neuronal death in focal cerebral ischemia and that pharmacological interventions aimed at neuroprotection may be usefully directed at cell cycle regulatory mechanisms.


Subject(s)
Brain Ischemia/metabolism , CDC2-CDC28 Kinases , Cell Cycle Proteins , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , Microtubule-Associated Proteins/metabolism , Neurons/metabolism , Proto-Oncogene Proteins , Tumor Suppressor Proteins , Animals , Brain Ischemia/pathology , Bromodeoxyuridine , Cell Cycle/physiology , Cell Death , Cell Hypoxia , Cells, Cultured , Cyclin D1/metabolism , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase Inhibitor p16/deficiency , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/metabolism , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Glucose/deficiency , Glucose/metabolism , In Situ Nick-End Labeling , Kinetin , Mice , Mice, Inbred Strains , Microtubule-Associated Proteins/deficiency , Neurons/pathology , Oxygen/metabolism , Protein Serine-Threonine Kinases/metabolism , Purines/pharmacology , Rats , Rats, Wistar
6.
Proc Natl Acad Sci U S A ; 98(10): 5874-9, 2001 May 08.
Article in English | MEDLINE | ID: mdl-11320217

ABSTRACT

Fibroblast growth factor-2 (FGF-2) promotes proliferation of neuroprogenitor cells in culture and is up-regulated within brain after injury. Using mice genetically deficient in FGF-2 (FGF-2(-/-) mice), we addressed the importance of endogenously generated FGF-2 on neurogenesis within the hippocampus, a structure involved in spatial, declarative, and contextual memory, after seizures or ischemic injury. BrdUrd incorporation was used to mark dividing neuroprogenitor cells and NeuN expression to monitor their differentiation into neurons. In the wild-type strain, hippocampal FGF-2 increased after either kainic acid injection or middle cerebral artery occlusion, and the numbers of BrdUrd/NeuN-positive cells significantly increased on days 9 and 16 as compared with the controls. In FGF-2(-/-) mice, BrdUrd labeling was attenuated after kainic acid or middle cerebral artery occlusion, as was the number of neural cells colabeled with both BrdUrd and NeuN. After FGF-2(-/-) mice were injected intraventricularly with a herpes simplex virus-1 amplicon vector carrying FGF-2 gene, the number of BrdUrd-labeled cells increased significantly to values equivalent to wild-type littermates after kainate seizures. These results indicate that endogenously synthesized FGF-2 is necessary and sufficient to stimulate proliferation and differentiation of neuroprogenitor cells in the adult hippocampus after brain insult.


Subject(s)
Brain Injuries/pathology , Fibroblast Growth Factor 2/physiology , Hippocampus/pathology , Animals , Brain Injuries/metabolism , Bromodeoxyuridine , Hippocampus/metabolism , Immunohistochemistry , Mice , Mice, Knockout
7.
J Neurosci ; 20(18): 6879-87, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-10995832

ABSTRACT

Cell death from spinal cord injury is mediated in part by apoptotic mechanisms involving downstream caspases (e.g., caspase-3). Upstream mechanisms may involve other caspases such as procaspase-8, a 55 kDa apical caspase, which we found constitutively expressed within spinal cord neurons along with Fas. As early as 1.5 hr after transient ischemia, activated caspase-8 (p18) and caspase-8 mRNA appeared within neurons in intermediate gray matter and in medial ventral horn. We also detected evidence for an increase in death receptor complex by co-immunoprecipitation using Fas and anti-procaspase-8 after ischemia. At early time points, Fas and p18 were co-expressed within individual neurons, as were activated caspase-8 and caspase-3. Moreover, we detected p18 in cells before procaspase-3 cleavage product (p20), suggesting sequential activation. The appearance of cytosolic cytochrome c and gelsolin cleavage after ischemia was consistent with mitochondrial release and caspase-3 activation, respectively. Numerous terminal deoxynucleotidyl transferase-mediated DNA nick end-labeling-positive neurons contained p18 or p20 (65 and 80%, respectively), thereby supporting the idea that cells undergoing cell death contain both processed caspases. Our data are consistent with the idea that transient spinal cord ischemia induces the formation of a death-inducing signaling complex, which may participate in caspase-8 activation and sequential caspase-3 cleavage. Death receptors as well as downstream caspases may be useful therapeutic targets for limiting the death of cells in spinal cord.


Subject(s)
Apoptosis , Neurons/metabolism , Spinal Cord Ischemia/metabolism , Spinal Cord/metabolism , fas Receptor/metabolism , Animals , Caspase 3 , Caspase 8 , Caspase 9 , Caspases/genetics , Caspases/metabolism , Cytochrome c Group/metabolism , Disease Models, Animal , Enzyme Precursors/genetics , Enzyme Precursors/metabolism , Female , Gelsolin/metabolism , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred C57BL , Neurons/pathology , RNA, Messenger/biosynthesis , Reperfusion , Signal Transduction , Spinal Cord/blood supply , Spinal Cord/pathology , Spinal Cord Ischemia/pathology
8.
J Cereb Blood Flow Metab ; 20(4): 709-17, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10779015

ABSTRACT

Nitric oxide, a product of nitric oxide synthase activity, relaxes vascular smooth muscle and elevates brain blood flow. We evaluated the importance of eNOS to cerebral blood flow augmentation after L-arginine infusion and increases in flow after eNOS upregulation in SV-129 mice. Blood flow was measured by laser-Doppler flowmetry before and after L-arginine infusion (450 mg/kg during a 15-minute period) or measured by 14C-iodoamphetamine indicator fractionation or 14C-iodoantipyrine tissue equilibration techniques. rCBF increased by 26% (laser Doppler flowmetry) after L-arginine infusion but did not change in mutant mice deficient in eNOS expression. After eNOS upregulation by chronic simvastatin treatment (2 mg/kg subcutaneously, daily for 14 days), L-arginine amplified and sustained the hyperemia (38%) and increased absolute brain blood flow from 86 +/- 7 to 119 +/- 10 mL/100 g per minute. Furthermore, pretreatment with simvastatin enhanced blood flow within ischemic brain tissue after middle cerebral artery occlusion. Together, these findings suggest that eNOS activity is critical for blood flow augmentation during acute L-arginine infusion, and chronic eNOS upregulation combined with L-arginine administration provides a novel strategy to elevate cerebral blood flow in the normal and ischemic brain.


Subject(s)
Arginine/pharmacology , Cerebrovascular Circulation/drug effects , Cerebrovascular Circulation/physiology , Nitric Oxide Synthase/physiology , Simvastatin/pharmacology , Animals , Arginine/blood , Arterial Occlusive Diseases/physiopathology , Cerebral Arteries , Drug Synergism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Mice , Mice, Inbred Strains/genetics , Nitric Oxide Synthase/deficiency , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Time Factors
9.
Stroke ; 31(1): 208-13, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10625739

ABSTRACT

BACKGROUND AND PURPOSE: Spinal cord ischemia with resulting paraplegia is a devastating complication of thoracoabdominal aortic surgery. Experimental models of spinal cord ischemia have been developed in primate, dog, pig, rabbit, and rat with variable reproducibility, but none has been developed in mouse. Because genetically engineered mice have become important to examine the impact of specific genes in ischemic pathophysiology, we sought to develop a reproducible mouse model of spinal cord ischemia. METHODS: C57BL/6NCrlBR mice were subjected to cross-clamping of the aortic arch, left subclavian artery, and internal mammary artery for 9 minutes (group A; n=8) or 11 minutes (group B; n=29) followed by reperfusion for 24 or 48 hours. Mean distal arterial blood pressure (left femoral artery) and lumbar (L1) spinal cord blood flow (laser-Doppler flowmetry) were measured for the duration of the procedure. The arterial blood supply of the spinal cord was visualized by intravascular perfusion of carbon black ink. We evaluated motor function in the hind limbs at 0, 1, 3, 6, and 24 hours after reperfusion using a rating scale of 0 (normal function) to 6 (total absence of movement). Spinal cord histopathology was evaluated after 24 and 48 hours of reperfusion by Luxol fast blue-hematoxylin and eosin. RESULTS: The vascular anatomy of the mouse and human spinal cord appeared similar in that blood was supplied by 1 anterior and 2 posterior spinal arteries and heterosegmental radicular arteries. During combined occlusion of aortic arch and left subclavian artery, mean distal arterial blood pressure dropped to 10+/-5 mm Hg, and spinal cord blood flow at the L1 level decreased to 27+/-7% of baseline. All animals recovered from anesthesia with acute paraplegia. Animals in the 9-minute group (group A) showed steady recovery of hind limb function over the ensuing 24 hours, whereas the majority (80%) in the 11-minute group (group B) remained paralyzed with maximum deficit throughout the postoperative period. Mortality was 0% and 21% in groups A and B, respectively. Maximal ischemic damage was observed at the lower thoracic and higher lumbar spinal levels in both groups. In group A (9 minutes), tissue damage was mild, affecting predominantly dorsal horns and intermediate gray matter, whereas ventral horns were minimally involved. All mice in group B (11 minutes) showed extensive gray matter lesions particularly involving dorsal horns and intermediate areas; in ventral horns, >50% of motor neurons died. White matter lesions were present in the most severely damaged cords only. CONCLUSIONS: Spinal cord ischemia caused by aortic arch plus left subclavian artery cross-clamping provides a mouse model useful for the study of spinal cord injury and of potential relevance to the complications following thoracoabdominal aortic surgery in humans.


Subject(s)
Disease Models, Animal , Spinal Cord Ischemia , Animals , Dogs , Humans , Mice , Mice, Inbred C57BL , Rabbits , Rats
10.
J Neurochem ; 73(3): 1212-21, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10461914

ABSTRACT

Sphingosine 1-phosphate or lysophosphatidic acid activation of guanosine-5'-O-(3-[35S]thio)triphosphate ([35S]GTPgammaS) binding to G proteins was studied by in vitro autoradiography in rat and guinea pig brain. The highest stimulation of [35S]GTPgammaS binding by sphingosine 1-phosphate was observed in the molecular layer of the cerebellum. Marked stimulation was observed in most forebrain areas, including neocortex and striatum. With the exception of the substantia gelatinosa and nucleus of the solitary tract, sphingosine 1-phosphate-enhanced binding was weaker in the brainstem and spinal cord. Lysophosphatidic acid-enhanced labeling was only observed in white matter areas. The G protein inhibitor 5'-p-fluorosulfonylbenzoyl guanosine completely inhibited lysophosphatidic acid-enhanced [35S]GTPgammaS binding but only partially sphingosine 1-phosphate-enhanced binding. N-Ethylmaleimide abolished binding stimulated by both agonists. Sphingosine 1-phosphate enhanced labeling by another GTP analogue (beta,gamma-imido[8-3H]guanosine-5'-triphosphate) similarly to that of [35S]GTPgammaS. Lysophosphatidic acid stimulated [35S]GTPgammaS binding in the olfactory bulb, glia limitans, and cortical subventricular zone of 1-day-old rats, whereas enhanced labeling was not observed in the latter area of 5-day-old rats. Sphingosine 1-phosphate stimulated binding in the cortical and striatal subventricular zones and olfactory bulb in 1- and 5-day-old rats. In the absence of radioligand for sphingosine 1-phosphate and lysophosphatidic acid receptors, [35S]GTPgammaS autoradiography provides a unique opportunity to study the spatial distribution, ontogeny, and coupling properties of these receptors.


Subject(s)
Brain/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Lysophospholipids/metabolism , Sphingosine/analogs & derivatives , Animals , Autoradiography , Binding Sites , Brain/drug effects , Enzyme Inhibitors/pharmacology , Ethylmaleimide/pharmacology , GTP-Binding Proteins/agonists , GTP-Binding Proteins/metabolism , Guanylyl Imidodiphosphate/metabolism , Guinea Pigs , Image Processing, Computer-Assisted , In Vitro Techniques , Male , Radioligand Assay , Rats , Rats, Sprague-Dawley , Sphingosine/metabolism , Sulfur Radioisotopes
11.
Neuropharmacology ; 38(7): 1043-53, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10428423

ABSTRACT

We studied the effects of PNU-109291 [(S)-(-)-1-[2-[4-(4-methoxyphenyl)-1-piperazinyl]ethyl]-N-methyl-isoc hroman-6-carboxamide], a receptor agonist showing 5000-fold selectivity for primate 5-HT1D versus 5-HT1B receptors (Ennis et al., J. Med. Chem. 41, 2180-2183), on dural neurogenic inflammation and on c-fos like immunoreactivity within trigeminal nucleus caudalis evoked by electrical and chemical activation of trigeminal afferents, respectively. Subcutaneous injection of PNU-109291 in male guinea pigs dose-dependently reduced dural extravasation of [125I]-labeled bovine serum albumin evoked by trigeminal ganglion stimulation with an IC50 of 4.2 nmol kg(-1). A dose of 73.3 nmol kg(-1) blocked the response completely. The selective 5-HT1B/1D receptor antagonist GR-127935 (> or = 2 micromol kg(-1) i.v.) prevented this effect. In addition, the number of c-fos immunoreactive cells within guinea pig trigeminal nucleus caudalis induced by chemical meningeal stimulation (intracisternally administered capsaicin) was reduced by more than 50% with PNU-109291 (> or = 122.2 nmol kg(-1) administered s.c. 45 min before and 15 min after capsaicin). These data indicate that the 5-HT1D receptor subtype plays a significant role in suppressing meningeal neurogenic inflammation and attenuating trigeminal nociception in these guinea pig models. Since 5-HT1D receptor mRNA and protein are expressed in trigeminal ganglia but not vascular smooth muscle, the 5-HT1D receptor subtype may become a useful therapeutic target for migraine and related headaches.


Subject(s)
Benzopyrans/pharmacology , Dura Mater/drug effects , Piperazines/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , Receptors, Serotonin/metabolism , Serotonin Receptor Agonists/pharmacokinetics , Trigeminal Nuclei/immunology , Trigeminal Nuclei/metabolism , Animals , Benzopyrans/pharmacokinetics , Binding, Competitive , Blood Pressure/drug effects , Blood Proteins/metabolism , Capsaicin/pharmacology , Dura Mater/immunology , Dura Mater/metabolism , Electric Stimulation , Guinea Pigs , Male , Oxadiazoles/pharmacology , Piperazines/pharmacokinetics , Receptor, Serotonin, 5-HT1D , Receptors, Serotonin/drug effects , Serotonin Antagonists/pharmacology , Serotonin Receptor Agonists/pharmacology , Trigeminal Nuclei/drug effects
12.
Br J Pharmacol ; 127(3): 623-30, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10401552

ABSTRACT

1. We examined the effects of the alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)/kainate receptor antagonists 6-cyano-7-nitroquinoxaline-2,3-dione (CNQX) and 1,2,3,4-tetrahydro-6-nitro-2,3-dioxo-benzol[f]quinoxaline-7-sulpho namide (NBQX), the kainate receptor antagonists gamma-(R-)-glutamylaminomethanesulphonic acid (GAMS) and 6,7,8,9-tetrahydro-5-nitro-1H-benz[g]indole-2,3-dione-3-oxime (NS-102), and the group III metabotropic glutamate receptor (mGluR) agonist 2-amino-4-phosphono-S-butanoic acid (L-AP4) on c-fos-like immunoreactivity (c-fos LI) in trigeminal caudalis (Sp5C), lateral reticular (LRt), medullary reticular (Md) and solitary tract (Sol) nuclei, after intracisternal injection of capsaicin in urethane anaesthetized Sprague-Dawley rats. 2. Few c-fos labelled cells were observed within Sp5C in capsaicin-vehicle treated animals. The number of positive c-fos cells increased by 17 fold after intracisternal capsaicin (5 nmol) administration. 3. Pretreatment with CNQX (0.02, 0.1, 0.6, 3 and 15 mg kg-1) or NBQX (0.01, 0.1 and 1 mg kg-1), administered intraperitoneally 15 min before capsaicin, significantly reduced labelled cells within Sp5C by a maximum of 45 and 34%, respectively. The number of c-fox LI cells within LRt, Md and Sol was not affected. Pretreatment with L-AP4 (1, 3 and 10 mg kg-1) decreased the number of Sp5C c-fos LI cells by a maximum of 30%, whereas GAMS (1 and 10 mg kg-1) and NS-102 (1 and 5 mg kg-1) did not show any significant effect. 4. These results suggest that blockade of AMPA receptors, but not kainate receptors, or the activation of group III mGluRs, decrease the response of Sp5C neurons to trigeminovascular activation. Thus, in addition to NMDA receptors, mGluRs and AMPA receptors may modulate cephalic pain and may provide a potential therapeutic target for antimigraine drugs.


Subject(s)
Capsaicin/pharmacology , Gene Expression Regulation , Genes, fos , Proto-Oncogene Proteins c-fos/biosynthesis , Receptors, Metabotropic Glutamate/physiology , Trigeminal Caudal Nucleus/physiology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Brain Stem/chemistry , Brain Stem/drug effects , Brain Stem/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Gene Expression Regulation/drug effects , Genes, fos/drug effects , Male , Neurons/chemistry , Neurons/drug effects , Neurons/metabolism , Quinoxalines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Kainic Acid/antagonists & inhibitors , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Trigeminal Caudal Nucleus/drug effects , Trigeminal Caudal Nucleus/metabolism
13.
Eur J Pharmacol ; 369(3): 271-7, 1999 Mar 26.
Article in English | MEDLINE | ID: mdl-10225363

ABSTRACT

A possible mechanism of action of antimigraine drugs such as sumatriptan is inhibition of the trigeminovascular pathway. Sumatriptan's effects might be mediated by 5-HT1B, 5-HT1D or 5-HT1F receptors. To establish the relative importance of these subtypes, we compared the effects of sumatriptan with those of a selective 5-HT1F receptor agonist (LY 344864) on c-fos protein expression in the trigeminal nucleus caudalis. c-fos expression was induced in urethane-anaesthetized rats by intracisternal capsaicin administration. Sumatriptan and LY 344864 decreased the number of capsaicin-induced c-fos-like immunoreactive cells within trigeminal nucleus caudalis (ID50 = 0.04 and 0.6 mg kg(-1)). The effect of sumatriptan, but not of LY 344864, was prevented by pretreatment with the antagonist SDZ 21-009, which displays high affinity for rat 5-HT1B receptors. LY 344864 appears to attenuate c-fos-like immunoreactivity via 5-HT1F receptors, while sumatriptan acts via 5-HT1B receptors. The fact that activation of 5-HT1F receptors is sufficient to modulate the activity of the trigeminal system suggests that this receptor may be a target for antimigraine drugs with improved safety profile.


Subject(s)
Carbazoles/pharmacology , Fluorobenzenes/pharmacology , Genes, fos/drug effects , Receptors, Serotonin/drug effects , Serotonin Receptor Agonists/pharmacology , Sumatriptan/pharmacology , Trigeminal Caudal Nucleus/drug effects , Adrenergic beta-Antagonists/pharmacology , Animals , Capsaicin/toxicity , Dose-Response Relationship, Drug , Drug Interactions , Gene Expression/drug effects , Male , Pindolol/analogs & derivatives , Pindolol/pharmacology , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley , Trigeminal Caudal Nucleus/metabolism
14.
Neuron ; 22(3): 581-91, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10197537

ABSTRACT

In order to determine the distribution and function of the 5-HT5A serotonin receptor subtype, we generated knockout mice lacking the 5-HT5A gene. Comparative autoradiography studies of brains of wild-type (wt) and 5-HT5A knockout (5A-KO) mice revealed the existence of binding sites with high affinity for [125I]LSD that correspond to 5-HT5A receptors and that are concentrated in the olfactory bulb, neocortex, and medial habenula. When exposed to novel environments, the 5A-KO mice displayed increased exploratory activity but no change in anxiety-related behaviors. In addition, the stimulatory effect of LSD on exploratory activity was attenuated in 5A-KO mice. These results suggest that 5-HT5A receptors modulate the activity of neural circuits involved specifically in exploratory behavior and suggest that some of the psychotropic effects of LSD may be mediated by 5-HT5A receptors.


Subject(s)
Exploratory Behavior/physiology , Hallucinogens/pharmacology , Lysergic Acid Diethylamide/pharmacology , Receptors, Serotonin/physiology , Animals , Animals, Newborn , Anxiety/physiopathology , Autoradiography , Immunohistochemistry , Male , Mice , Mice, Knockout , Motor Activity/drug effects , Motor Activity/physiology , Receptors, Serotonin/genetics , Reflex, Startle/drug effects , Reflex, Startle/genetics , Reflex, Startle/physiology
15.
Pain ; 76(1-2): 239-48, 1998 May.
Article in English | MEDLINE | ID: mdl-9696479

ABSTRACT

The effect of the N-methyl-D-aspartate (NMDA) receptor antagonist (5R, 10S)-(+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclo-hepten-5,10-i mine hydrogen maleate (MK-801) was examined on c-fos-like immunoreactivity (c-fos-LI) in urethane-anesthetized Sprague-Dawley rats using a polyclonal antibody. C-fos, an indicator of neuronal activation, was assessed within the trigeminal nucleus caudalis (TNC), area postrema. lateral reticular and solitary tract nuclei 2 h after intracisternal injection of capsaicin. C-fos-positive cells were counted at three representative levels corresponding to obex, -2.05 mm and -6.45 mm in 18 tissue sections (50 microm). A weighted average was obtained reflecting total brainstem expression within lamina I, II of TNC using a recently validated method. Capsaicin (0.1, 1, 5, 10 and 15 nmol) caused a dose-dependent labeling of cells in lamina I, II at obex similar to that previously reported after intracisternal blood or carrageenin administration in rats and guinea pigs. MK-801 (0.3, 1 and 3 mg/kg) administered i.p. 30 min before capsaicin (5 nmol in 100 microl artificial CSF) reduced significantly and dose-dependently (12%, 36% and 47%, respectively) the c-fos-LI cells in TNC at each level from rostral to caudal but not in solitary tract, area postrema and lateral reticular nuclei, and for unexplained reasons, increased c-fos-LI within the inferior olive. These results suggest that NMDA receptors provide a potential therapeutic target for cephalic pain (e.g. migraine) due to trigeminovascular activation from meningeal afferents.


Subject(s)
Capsaicin/antagonists & inhibitors , Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Gene Expression Regulation/drug effects , Genes, fos/drug effects , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Trigeminal Nuclei/metabolism , Animals , Blood Pressure/drug effects , Capsaicin/pharmacology , Cell Count/drug effects , Depression, Chemical , Heart Rate/drug effects , Immunohistochemistry , Male , Rats , Rats, Sprague-Dawley , Time Factors , Trigeminal Nuclei/drug effects
17.
Am J Physiol ; 274(2): H411-5, 1998 02.
Article in English | MEDLINE | ID: mdl-9486242

ABSTRACT

We evaluated the effects of superfusing 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ), eNOS null (B) an inhibitor of soluble guanylyl cyclase, and 7-nitroindazole sodium (7-NI), a selective neuronal nitric oxide synthase (nNOS) inhibitor, on the acetylcholine (ACh) response in endothelial NOS (eNOS) null mice. Pial arteriolar diameter was measured by intravital microscopy through a closed cranial window under alpha-chloralose anesthesia. NOS activity was measured by [3H]arginine-to-[3H]citrulline conversion in subjacent cortex in vitro. The density and distribution of muscarinic receptors in the brain were determined by quantitative [3H]quinuclidinyl benzilate autoradiography and did not differ between the eNOS mutants and wild-type mice. ACh superfusion (1 and 10 microM) dose dependently dilated pial arterioles in eNOS null and wild-type mice. ODQ (10 microM) attenuated ACh-induced dilation in both eNOS mutants (41% decrease at 10 microM ACh, P < 0.01, n = 6) and wild-type strains (n = 5 per group). By contrast, topical superfusion of 7-NI (100 microM) attenuated the ACh response in eNOS mutants only (66%, P < 0.05, and 25% decrease, P < 0.05, at 1 and 10 microM ACh, respectively). Our findings suggest that nNOS-guanosine 3',5'-cyclic monophosphate (cGMP)-dependent pathways dilate pial arterioles by compensatory mechanisms after eNOS gene disruption.


Subject(s)
Acetylcholine/pharmacology , Cyclic GMP/pharmacology , Endothelium, Vascular/enzymology , Nitric Oxide Synthase/physiology , Pia Mater/blood supply , Vasodilation/drug effects , Animals , Arginine/metabolism , Arterioles/drug effects , Arterioles/physiology , Autoradiography , Brain/metabolism , Citrulline/metabolism , Enzyme Inhibitors/pharmacology , Indazoles/pharmacology , Mice , Mice, Knockout , Neurons/enzymology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/genetics , Quinuclidinyl Benzilate/metabolism , Receptors, Muscarinic/analysis , Receptors, Muscarinic/metabolism
18.
J Cereb Blood Flow Metab ; 18(3): 238-47, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9498840

ABSTRACT

Inhibitors of apoptosis and of excitotoxic cell death reduce brain damage after transient and permanent middle cerebral artery occlusion. We compared the neuroprotective effects of two caspase family inhibitors with the N-methyl-D-aspartate receptor antagonist (+)-MK-801 hydrogen maleate (MK-801) in a newly characterized cycloheximide-sensitive murine model of transient middle cerebral artery occlusion (30 minutes) in which apoptotic cell death is prominent. Ischemic infarction, undetected by 2,3,5-triphenyltetrazolium chloride staining at 24-hour reperfusion, featured prominently in the striatum at 72 hours and 7 days on hematoxylin-eosin-stained sections. Markers of apoptosis, such as oligonucleosomal DNA damage (laddering) and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick-end labeling (TUNEL)-positive cells first appeared at 24 hours and increased significantly at 72 hours and 7 days after reperfusion. The TUNEL-labeled cells were mostly neurons and stained negative for glial (GFAP, glial fibrillary acid protein) and leukocyte specific markers (CD-45). The caspase inhibitors, N-benzyloxycarbonyl-Val-Ala-Asp-fluoromethyl ketone (z-VAD.FMK; 120 ng intracerebroventricularly) or N-benzyloxycarbonyl-Asp-Glu-Val-Asp-fluoromethyl ketone (z-DEVD.FMK; 480 ng intracerebroventricularly) decreased infarct size and neurologic deficits when administered 6 hours after reperfusion. The extent of protection was greater than in models of more prolonged ischemia or after permanent occlusion, and the therapeutic window was extended from 0 to 1 hours after 2-hour middle cerebral artery occlusion to at least 6 hours after brief ischemia. Also, z-VAD.FMK and z-DEVD.FMK treatment decreased oligonucleosomal DNA damage (DNA laddering) as assessed by quantitative autoradiography after gel electrophoresis. By contrast, MK-801 protected brain tissue only when given before ischemia (3 mg/kg intraperitoneally), but not at 3 or 6 hours after reperfusion. Despite a decrease in infarct size after MK-801 pretreatment, the amount of DNA laddering did not decrease 72 hours after reperfusion, thereby suggesting a mechanism distinct from inhibition of apoptosis. Hence, 30 minutes of reversible ischemia augments apoptotic cell death, which can be attenuated by delayed z-VAD.FMK and z-DEVD.FMK administration with preservation of neurologic function. By contrast, the therapeutic window for MK-801 does not extend beyond the time of occlusion, probably because its primary mechanism of action does not block the development of apoptotic cell death.


Subject(s)
Amino Acid Chloromethyl Ketones/administration & dosage , Apoptosis/drug effects , Brain Ischemia/pathology , Caspases , Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/administration & dosage , Neurons/pathology , Oligopeptides/administration & dosage , Animals , Brain Ischemia/drug therapy , Brain Ischemia/enzymology , Caspase 1 , Caspase 3 , Cell Death/drug effects , Cerebral Infarction/prevention & control , Injections, Intraventricular , Male , Mice , Neurons/enzymology
19.
Ann N Y Acad Sci ; 861: 85-90, 1998 Dec 15.
Article in English | MEDLINE | ID: mdl-9928243

ABSTRACT

Putative 5-ht5 receptor binding sites were visualized by in vitro autoradiography using [125I]LSD (in the presence of clozapine and spiperone) or [3H]5-carboxamidotryptamine (in the presence 8-OH-DPAT, GR127935 and spiperone). Under these conditions, no [3H]5-carboxamidotryptamine labeling was detected in the brain of mice lacking the gene encoding the putative 5-ht5a receptor (knockout mice), whereas intermediate densities of binding sites were seen in the olfactory bulb and neocortex of wild-type mice. [125I]LSD labeled the same areas as [3H]5-carboxamidotryptamine in wild-type mice. High densities of [125I]LSD binding sites were observed in the medial habenula of wild type and knockout mice. 5-CT competed for [125I]LSD binding sites with an affinity of 2 nM in the olfactory bulb and neocortex of wild-type mice and an affinity of 30 nM in the habenula of knockout mice, suggesting that habenular labeling might be accounted for by putative 5-ht5b receptors. In the presence of 5'-guanylylimidodiphosphate, 5-CT displaced [125I]LSD from putative 5-ht5a and 5-ht5b sites with a 6-times and 3-times lower affinity, respectively, suggesting that both receptor subtypes are coupled to G proteins in brain. We also studied the inhibitory effect of 5-CT on dural neurogenic inflammation in knockout mice. In wild type mice, 3 ng/kg 5-CT inhibited dural protein extravasation by 60%. A similar effect was observed in knockout mice, even in the presence of the 5-HT1B receptor antagonist GR127935. These results suggest that the inhibitory effects of 5-CT are not mediated by a site with the characteristics of the putative 5-ht5 receptor.


Subject(s)
Blood Proteins/metabolism , Brain/physiology , Receptors, Serotonin/physiology , Trigeminal Ganglion/physiology , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Animals , Autoradiography , Binding Sites , Brain/drug effects , Crosses, Genetic , Female , Lysergic Acid Diethylamide/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Specificity , Oxadiazoles/pharmacology , Piperazines/pharmacology , Receptors, Serotonin/deficiency , Receptors, Serotonin/genetics , Serotonin/analogs & derivatives , Serotonin/metabolism , Serotonin Antagonists/pharmacology , Trigeminal Ganglion/drug effects , Tritium
20.
J Cereb Blood Flow Metab ; 17(11): 1143-51, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9390645

ABSTRACT

Poly(ADP-ribose)polymerase (PARP, EC 2.4.2.30), an abundant nuclear protein activated by DNA nicks, mediates cell death in vitro by nicotinamide adenine dinucleotide (NAD) depletion after exposure to nitric oxide. The authors examined whether genetic deletion of PARP (PARP null mice) or its pharmacologic inhibition by 3-aminobenzamide (3-AB) attenuates tissue injury after transient cerebral ischemia. Twenty-two hours after reperfusion following 2 hours of filamentous middle cerebral artery occlusion, ischemic injury was decreased in PARP-/- and PARP+/- mice compared with PARP+/+ litter mates, and also was attenuated in 129/SV wild-type mice after 3-AB treatment compared with controls. Infarct sparing was accompanied by functional recovery in PARP-/- and 3-AB-treated mice. Increased poly(ADP-ribose) immunostaining observed in ischemic cell nuclei 5 minutes after reperfusion was reduced by 3-AB treatment. Levels of NAD--the substrate of PARP--were reduced 2 hours after reperfusion and were 35% of contralateral levels at 24 hours. The decreases were attenuated in PARP-/- mice and in 3-AB-treated animals. Poly(ADP-ribose)polymerase cleavage by caspase-3 (CPP-32) has been proposed as an important step in apoptotic cell death. Markers of apoptosis, such as oligonucleosomal DNA damage, total DNA fragmentation, and the density of terminal deoxynucleotidyl transferase dUTP nick-end-labelled (TUNEL +) cells, however, did not differ in ischemic brain tissue of PARP-/- mice or in 3-AB-treated animals versus controls, although there were differences in the number of TUNEL-stained cells reflecting the decrease in infarct size. Thus, ischemic brain injury activates PARP and contributes to cell death most likely by NAD depletion and energy failure, although the authors have not excluded a role for PARP in apoptotic cell death at earlier or later stages in ischemic cell death. Inhibitors of PARP activation could provide a potential therapy in acute stroke.


Subject(s)
Brain Damage, Chronic/etiology , Brain Ischemia/complications , Poly(ADP-ribose) Polymerases/metabolism , Animals , Benzamides/pharmacology , Brain/metabolism , Brain Ischemia/genetics , Brain Ischemia/metabolism , Cerebral Infarction/etiology , Cerebral Infarction/pathology , DNA Damage/physiology , DNA Fragmentation/physiology , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Male , Mice , Mice, Inbred Strains , NAD/metabolism , Poly Adenosine Diphosphate Ribose/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...