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










Publication year range
1.
Physiol Behav ; 105(3): 669-75, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22019828

ABSTRACT

Brain substance P and its receptor (neurokinin-1, NK1) have a widespread brain distribution and are involved in an important number of behavioural and physiological responses to emotional stimuli. However, the role of NK1 receptors in the consequences of exposure to chronic stress has not been explored. The present study focused on the role of these receptors in the hypothalamic-pituitary-adrenal (HPA) response to daily repeated restraint stress (evaluated by plasma corticosterone levels), as well as on the effect of this procedure on anxiety-like behaviour, spatial learning and memory in the Morris water maze (MWM), a hippocampus-dependent task. Adult null mutant NK1-/- mice, with a C57BL/6J background, and the corresponding wild-type mice showed similar resting corticosterone levels and, also, did not differ in corticosterone response to a first restraint. Nevertheless, adaptation to the repeated stressor was faster in NK1-/- mice. Chronic restraint modestly increased anxiety-like behaviour in the light-dark test, irrespective of genotype. Throughout the days of the MWM trials, NK1-/- mice showed a similar learning rate to that of wild-type mice, but had lower levels of thigmotaxis and showed a better retention in the probe trial. Chronic restraint stress did not affect these variables in either genotype. These results indicate that deletion of the NK1 receptor does not alter behavioural susceptibility to chronic repeated stress in mice, but accelerates adaptation of the HPA axis. In addition, deletion may result in lower levels of thigmotaxis and improved short-term spatial memory, perhaps reflecting a better learning strategy in the MWM.


Subject(s)
Behavior, Animal/physiology , Corticosterone/blood , Receptors, Neurokinin-1/deficiency , Stress, Psychological , Adaptation, Ocular , Animals , Chi-Square Distribution , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Radioimmunoassay/methods , Restraint, Physical/methods , Stress, Psychological/blood , Stress, Psychological/metabolism , Stress, Psychological/physiopathology , Time Factors
2.
Circ Res ; 95(10): 1027-34, 2004 Nov 12.
Article in English | MEDLINE | ID: mdl-15499026

ABSTRACT

Myogenic constriction describes the innate ability of resistance arteries to constrict in response to elevations in intraluminal pressure and is a fundamental determinant of peripheral resistance and, hence, organ perfusion and systemic blood pressure. However, the receptor/cell-type that senses changes in pressure on the blood vessel wall and the pathway that couples this to constriction of vascular smooth muscle remain unclear. In this study, we show that elevation of intraluminal transmural pressure of mesenteric small arteries in vitro results in a myogenic response that is profoundly suppressed following ablation of sensory C-fiber activity (using in vitro capsaicin desensitization resulted in 72.8+/-10.3% inhibition, n=8; P<0.05). Activation of C-fiber nerve endings by pressure was attributable to stimulation of neuronal vanilloid receptor, TRPV1, because blockers of this channel, capsazepine (71.9+/-11.1% inhibition, n=9; P<0.001) and ruthenium red (46.1+/-11.7% inhibition, n=4; P<0.05), suppressed the myogenic constriction. In addition, this C-fiber dependency is likely related to neuropeptide substance P release and activity because blockade of tachykinin NK1 receptors (66.3+/-13.7% inhibition, n=6; P<0.001), and not NK2 receptors (n=4, NS), almost abolished the myogenic response. Previous studies support a role for 20-hydroxyeicosatetraenoic acid (20-HETE) in myogenic constriction responses; herein, we show that 20-HETE-induced constriction of mesenteric resistance arteries is blocked by capsazepine. Together, these results suggest that elevation of intraluminal pressure is associated with generation of 20-HETE that, in turn, activates TRPV1 on C-fiber nerve endings resulting in depolarization of nerves and consequent vasoactive neuropeptide release. These findings identify a novel mechanism contributing to Bayliss' myogenic constriction and highlights an alternative pathway that may be targeted in the therapeutics of vascular disease, such as hypertension, where enhanced myogenic constriction plays a role in the pathogenesis.


Subject(s)
Capsaicin/analogs & derivatives , Ion Channels/physiology , Mesenteric Arteries/physiology , Models, Cardiovascular , Models, Neurological , Nerve Fibers, Unmyelinated/physiology , Splanchnic Circulation/physiology , Vascular Resistance/physiology , Vasoconstriction/physiology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Animals , CHO Cells , Capsaicin/pharmacology , Capsaicin/toxicity , Cation Transport Proteins/antagonists & inhibitors , Cricetinae , Endothelium, Vascular/physiology , Gadolinium/pharmacology , Ganglia, Sympathetic/drug effects , Guanethidine/pharmacology , Hydroxyeicosatetraenoic Acids/pharmacology , Ion Channels/drug effects , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/innervation , Mice , Mice, Knockout , Nerve Fibers, Unmyelinated/drug effects , Nociceptors/drug effects , Nociceptors/physiology , Peptides, Cyclic/pharmacology , Piperidines/pharmacology , Pressure , Quinuclidines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Neurokinin-1/deficiency , Receptors, Neurokinin-1/genetics , Ruthenium Red/pharmacology , Sodium Channel Blockers/pharmacology , Splanchnic Circulation/drug effects , Stress, Mechanical , Sympathectomy, Chemical , TRPV Cation Channels , Tetrodotoxin/pharmacology , Vascular Resistance/drug effects , Vasoconstriction/drug effects
3.
Pain ; 111(1-2): 169-80, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15327821

ABSTRACT

Patients with metastatic breast, lung or prostate cancer frequently have significant bone cancer pain. In the present report we address, in a single in vivo mouse model, the effects the bisphosphonate alendronate has on bone cancer pain, bone remodeling and tumor growth and necrosis. Following injection and confinement of green fluorescent protein-transfected murine osteolytic tumor cells into the marrow space of the femur of male C3H/HeJ mice, alendronate was administered chronically from the time the tumor was established until the bone cancer pain became severe. Alendronate therapy reduced ongoing and movement-evoked bone cancer pain, bone destruction and the destruction of sensory nerve fibers that innervate the bone. Whereas, alendronate treatment did not change viable tumor burden, both tumor growth and tumor necrosis increased. These data emphasize that it is essential to utilize a model where pain, skeletal remodeling and tumor growth can be simultaneously assessed, as each of these can significantly impact patient quality of life and survival.


Subject(s)
Alendronate/pharmacology , Bone Neoplasms/drug therapy , Osteolysis/drug therapy , Pain/drug therapy , Sarcoma/drug therapy , Activating Transcription Factor 3 , Animals , Behavior, Animal , Biomarkers, Tumor , Bone Neoplasms/complications , Bone Neoplasms/pathology , Male , Mice , Mice, Inbred C3H , Necrosis , Osteoclasts/drug effects , Osteoclasts/pathology , Osteolysis/etiology , Osteolysis/pathology , Pain/etiology , Pain/pathology , Sarcoma/complications , Sarcoma/pathology , Transcription Factors/metabolism
4.
Eur J Pharmacol ; 492(1): 41-8, 2004 May 10.
Article in English | MEDLINE | ID: mdl-15145704

ABSTRACT

We recently demonstrated that mice lacking the gene for substance P (neurokinin 1) receptors (NK1-/-) show improved cortical dialysate serotonin (5-HT) responses to paroxetine [J. Neurosci. 21 (2001) 8188]. To test for changes that may involve the 5-HT transporter (5-HTT) in these mutant mice, in vivo/in vitro studies were performed. Autoradiographic quantification of 5-HTT was performed: [3H]citalopram binding did not reveal any modification of 5-HT binding sites in the dorsal raphe nucleus (DRN) of wild-type NK1+/+ control and mutant NK1-/- mice. These results were further confirmed by 5-HTT mRNA quantification using competitive reverse transcription and polymerase chain reaction (RT-PCR) assay, which showed similar messenger levels in the DRN of both mice genotypes. The functional status of 5-HTT in vivo was tested by using the zero net flux method of quantitative microdialysis in two serotonergic nerve terminal regions, the frontal cortex and ventral hippocampus, of wild-type NK1+/+ and NK1-/- mice. Neither basal extracellular 5-HT levels nor the 5-HT extraction fraction of the probe (Ed an index of 5-HT uptake in vivo) differed between wild-type and mutant mice in the two brain regions studied. These results suggest that no compensatory response to the constitutive deletion of the tachykinin NK1 receptor involving changes in the activity of the selective 5-HT transporter occurred in the DRN, frontal cortex and ventral hippocampus in mice.


Subject(s)
Membrane Glycoproteins/metabolism , Membrane Transport Proteins/metabolism , Nerve Tissue Proteins/metabolism , Raphe Nuclei/metabolism , Receptors, Neurokinin-1/genetics , Serotonin/metabolism , Animals , Autoradiography , Citalopram/pharmacology , Extracellular Fluid/metabolism , Frontal Lobe/drug effects , Frontal Lobe/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Membrane Glycoproteins/genetics , Membrane Transport Proteins/genetics , Mice , Mice, Knockout , Microdialysis , Nerve Tissue Proteins/genetics , RNA, Messenger/analysis , RNA, Messenger/metabolism , Raphe Nuclei/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Serotonin/analysis , Serotonin Plasma Membrane Transport Proteins , Selective Serotonin Reuptake Inhibitors/pharmacology
5.
J Neurochem ; 89(1): 54-63, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15030389

ABSTRACT

Abstract Substance P antagonists of the neurokinin-1 receptor type (NK1) are gaining growing interest as new antidepressant therapies. It has been postulated that these drugs exert this putative therapeutic effect without direct interactions with serotonin (5-HT) neurones. Our recent microdialysis experiment performed in NK1 receptor knockout mice suggested evidence of changes in 5-HT neuronal function (Froger et al. 2001). The aim of the present study was to evaluate the effects of coadministration of the selective 5-HT reuptake inhibitor (SSRI) paroxetine with a NK1 receptor antagonist (GR205171 or L733060), given either intraperitoneally (i.p.) or locally into the dorsal raphe nucleus, on extracellular levels of 5-HT ([5-HT]ext) in the frontal cortex and the dorsal raphe nucleus using in vivo microdialysis in awake, freely moving mice. The systemic or intraraphe administration of a NK1 receptor antagonist did not change basal cortical [5-HT]ext in mice. A single systemic dose of paroxetine (4 mg/kg; i.p.) resulted in a statistically significant increase in [5-HT]ext with a larger extent in the dorsal raphe nucleus (+ 138% over basal AUC values), than in the frontal cortex (+ 52% over basal AUC values). Co-administration of paroxetine (4 mg/kg; i.p.) with the NK1 receptor antagonists, GR205171 (30 mg/kg; i.p.) or L733060 (40 mg/kg; i.p.), potentiated the effects of paroxetine on cortical [5-HT]ext in wild-type mice, whereas GR205171 (30 mg/kg; i.p.) had no effect on paroxetine-induced increase in cortical [5-HT]ext in NK1 receptor knock-out mice. When GR205171 (300 micro mol/L) was perfused by 'reverse microdialysis' into the dorsal raphe nucleus, it potentiated the effects of paroxetine on cortical [5-HT]ext, and inhibited paroxetine-induced increase in [5-HT]ext in the dorsal raphe nucleus. Finally, in mice whose 5-HT transporters were first blocked by a local perfusion of 1 micro mol/L of citalopram into the frontal cortex, a single dose of paroxetine (4 mg/kg i.p.) decreased cortical 5-HT release, and GR205171 (30 mg/kg i.p.) reversed this effect. The present findings suggest that NK1 receptor antagonists, when combined with a SSRI, augment 5-HT release by modulating substance P/5-HT interactions in the dorsal raphe nucleus.


Subject(s)
Frontal Lobe/metabolism , Neurokinin-1 Receptor Antagonists , Piperidines/pharmacology , Raphe Nuclei/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , Serotonin/metabolism , Tetrazoles/pharmacology , Animals , Drug Administration Routes , Extracellular Fluid/chemistry , Extracellular Fluid/metabolism , Frontal Lobe/drug effects , Injections, Intraperitoneal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microdialysis , Paroxetine/pharmacology , Perfusion , Raphe Nuclei/drug effects , Receptors, Neurokinin-1/metabolism , Serotonin/analysis
6.
Eur J Neurosci ; 19(5): 1133-40, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15016072

ABSTRACT

We have generated embryonic stem (ES) cells and transgenic mice with green fluorescent protein (GFP) inserted into the Pitx3 locus via homologous recombination. In the central nervous system, Pitx3-directed GFP was visualized in dopaminergic (DA) neurons in the substantia nigra and ventral tegmental area. Live primary DA neurons can be isolated by fluorescence-activated cell sorting from these transgenic mouse embryos. In culture, Pitx3-GFP is coexpressed in a proportion of ES-derived DA neurons. Furthermore, ES cell-derived Pitx3-GFP expressing DA neurons responded to neurotrophic factors and were sensitive to DA-specific neurotoxin N-4-methyl-1, 2, 3, 6-tetrahydropyridine. We anticipate that the Pitx3-GFP ES cells could be used as a powerful model system for functional identification of molecules governing mDA neuron differentiation and for preclinical research including pharmaceutical drug screening and transplantation. The Pitx3 knock-in mice, on the other hand, could be used for purifying primary neurons for molecular studies associated with the midbrain-specific DA phenotype at a level not previously feasible. These mice would also provide a useful tool to study DA fate determination from embryo- or adult-derived neural stem cells.


Subject(s)
Dopamine/biosynthesis , Luminescent Proteins/biosynthesis , Mesencephalon/metabolism , Neurons/metabolism , Stem Cells/metabolism , Animals , Cells, Cultured , Dopamine/genetics , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental/physiology , Green Fluorescent Proteins , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Luminescent Proteins/genetics , Male , Mesencephalon/cytology , Mesencephalon/embryology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/cytology , Pregnancy , Stem Cells/cytology , Transcription Factors/biosynthesis , Transcription Factors/genetics
7.
Brain Res ; 1002(1-2): 1-10, 2004 Mar 26.
Article in English | MEDLINE | ID: mdl-14988027

ABSTRACT

Antidepressants are widely prescribed in the treatment of depression, although the mechanism of how they exert their therapeutic effects is poorly understood. To shed further light on their mode of action, we have attempted to identify a common proteomic signature in guinea pig brains after chronic treatment with two different antidepressants. Both fluoxetine and the substance P receptor (NK(1)R) antagonist (SPA) L-000760735 altered cortical expression of multiple heat shock protein 60 forms along with neurofilaments and related proteins that are critical determinants of synaptic structure and function. Analysis of NK(1)R-/- mice showed similar alterations of neurofilaments confirming the specificity of the effects observed with chronic NK(1)R antagonist treatment. To determine if these changes were associated with structural modification of synapses, we carried out electron microscopic analysis of cerebral cortices from fluoxetine-treated guinea pigs. This showed an increase in the percentage of synapses with split postsynaptic densities (PSDs), a phenomenon that is characteristic of activity-dependent synaptic rearrangement. These findings suggest that cortical alterations of the neurofilament pathway and increased synaptic remodeling are associated with the mechanism of these two antidepressant drug treatments and may contribute to their psychotherapeutic actions.


Subject(s)
Antidepressive Agents/pharmacology , Fluoxetine/pharmacology , Neurofilament Proteins/ultrastructure , Neurokinin-1 Receptor Antagonists , Synapses/drug effects , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/ultrastructure , Guinea Pigs , Heat-Shock Proteins/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurofilament Proteins/biosynthesis , Receptors, Neurokinin-1/biosynthesis , Receptors, Neurokinin-1/deficiency , Synapses/metabolism , Synapses/ultrastructure
8.
Eur J Neurosci ; 18(7): 1828-36, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14622216

ABSTRACT

It has previously been shown that chronic treatment with antidepressant drugs increases neurogenesis and levels of brain-derived neurotrophic factor in the hippocampus. These changes have been correlated with changes in learning and long-term potentiation and may contribute to the therapeutic efficacy of antidepressant drug treatment. Recently, antagonists at the neurokinin-1 receptor, the preferred receptor for the neuropeptide substance P, have been shown to have antidepressant activity. Mice with disruption of the neurokinin-1 receptor gene are remarkably similar both behaviourally and neurochemically to mice maintained chronically on antidepressant drugs. We demonstrate here that there is a significant elevation of neurogenesis but not cell survival in the hippocampus of neurokinin-1 receptor knockout mice. Neurogenesis can be increased in wild-type but not neurokinin-1 receptor knockout mice by chronic treatment with antidepressant drugs which preferentially target noradrenergic and serotonergic pathways. Hippocampal levels of brain-derived neurotrophic factor are also two-fold higher in neurokinin-1 receptor knockout mice, whereas cortical levels are similar. Finally, we examined hippocampus-dependent learning and memory but found no clear enhancement in neurokinin-1 receptor knockout mice. These data argue against a simple correlation between increased levels of neurogenesis or brain-derived neurotrophic factor and mnemonic processes in the absence of increased cell survival. They support the hypothesis that increased neurogenesis, perhaps accompanied by higher levels of brain-derived neurotrophic factor, may contribute to the efficacy of antidepressant drug therapy.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Cell Division/physiology , Mice, Knockout/metabolism , Receptors, Neurokinin-1/metabolism , Animals , Animals, Newborn , Antidepressive Agents/administration & dosage , Behavior, Animal/drug effects , Blotting, Western , Bromodeoxyuridine/pharmacokinetics , Cell Count , Cell Division/drug effects , Cell Survival/drug effects , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Conditioning, Psychological , Fear/drug effects , Genotype , Hippocampus , Immobilization , Immunohistochemistry , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Phosphorus Isotopes/metabolism , Radiation-Sensitizing Agents/pharmacokinetics , Reaction Time/drug effects , Receptors, Neurokinin-1/deficiency , Receptors, Neurokinin-1/genetics , Thymidine/metabolism , Time Factors , Tritium/metabolism
9.
Neuroreport ; 14(17): 2189-92, 2003 Dec 02.
Article in English | MEDLINE | ID: mdl-14625445

ABSTRACT

We sought to characterise the contribution of the neuropeptide substance P to the outcome of two models of footpad inflammation of differing severity. In an intense inflammatory model produced by intra-plantar Mycobacterium tuberculosus (10 mg/ml) substantial reductions in footpad swelling, histological outcome and mechanical hyperalgesia were observed from early time points in mice lacking the neurokin-1 receptor for substance P compared with wild-type controls. Conversely, in a less intense model (M. tuberculosus 1 mg/ml) no differences were observed other than for a reduction in mechanical hyperalgesia at later time points (day 9 onwards). The results point to a previously unrecognised influence of substance P on peripheral tissue injury and the maintenance of hyperalgesia during more severe or more chronic phases of inflammatory disease.


Subject(s)
Arthritis, Experimental/genetics , Hyperalgesia/genetics , Inflammation/genetics , Receptors, Neurokinin-1/deficiency , Receptors, Neurokinin-1/genetics , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/prevention & control , Female , Hyperalgesia/prevention & control , Inflammation/prevention & control , Male , Mice , Mice, Knockout
11.
J Neurosci ; 23(23): 8271-80, 2003 Sep 10.
Article in English | MEDLINE | ID: mdl-12967989

ABSTRACT

Mice lacking the neurokinin-1 (NK1) receptor, the preferred receptor for the neuropeptide substance P (SP), do not show many of the behaviors associated with morphine reward. To identify the areas of the brain that might contribute to this effect, we assessed the behavioral effects of ablation of neurons expressing the NK1 receptor in specific regions of the mouse brain using the neurotoxin substance P-saporin. In a preliminary investigation, bilateral ablation of these neurons from the amygdala, but not the nucleus accumbens and dorsomedial caudate putamen, brought about reductions in morphine reward behavior. Subsequently, the effect of ablation of these neurons in the amygdala on anxiety behavior was assessed using the elevated plus maze (EPM), before conditioned place preference (CPP), and locomotor responses to morphine were measured. Loss of NK1 receptor-expressing neurons in the amygdala caused an increase in anxiety-like behavior on the EPM. It also brought about a reduction in morphine CPP scores and the stimulant effect of acute morphine administration relative to saline controls, without affecting CPP to cocaine. NK1 receptor-expressing neurons in the mouse amygdala therefore modulate morphine reward behaviors. These observations mirror those observed in NK1 receptor knock-out (NK1-/-) mice and suggest that the amygdala is an important area for the effects of SP and the NK1 receptor in the motivational properties of opiates, as well as the control of behaviors related to anxiety.


Subject(s)
Amygdala/metabolism , Anxiety/genetics , Morphine/pharmacology , Neurons/metabolism , Receptors, Neurokinin-1/metabolism , Reward , Amygdala/cytology , Amygdala/drug effects , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Cocaine/pharmacology , Conditioning, Psychological , Crosses, Genetic , Drug Administration Routes , Illicit Drugs/pharmacology , Immunotoxins/pharmacology , Male , Maze Learning/drug effects , Mice , Mice, Inbred Strains , Mice, Knockout , Motor Activity/drug effects , Narcotics/pharmacology , Neurons/cytology , Neurons/drug effects , Nucleus Accumbens/cytology , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Receptors, Neurokinin-1/deficiency , Receptors, Neurokinin-1/genetics , Ribosome Inactivating Proteins, Type 1 , Saporins , Spatial Behavior/drug effects , Substance P/analogs & derivatives , Substance P/pharmacology
12.
Proteomics ; 3(7): 1162-71, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12872217

ABSTRACT

This paper describes the use of fluorescence two-dimensional differential in-gel electrophoresis in a multiplex analysis of two distinct proteomes. As a model system, cerebral cortex tissues were analyzed from neurokinin1 receptor knockout (NK(1)R-/-) and wild type (NK(1)R+/+) mice in an attempt to identify molecular pathways involved in the function of this protein. Paired NK(1)R-/- and NK(1)R+/+ samples were labeled with fluorescent Cy3 and Cy5 dyes and electrophoresed on the same two-dimensional gels. Scanning the gels at wavelengths specific for each dye revealed the two different proteomes which were overlaid and the differences in abundance of specific protein spots were determined by the Amersham Biosciences DeCyder Differential In-gel Analysis software. A Cy2-labeled sample pool was co-electrophoresed with all Cy3- and Cy5-labeled sample pairs as an internal standard providing a link for inter-gel comparisons and for more robust statistical analysis of the data. Eight spots were found to be upregulated and two downregulated in the NK(1)R-/- mice compared to NK(1)R+/+ controls. Matrix assisted laser desorption/ionisation-time of flight (MALDI-TOF) mass fingerprinting was used to identify the proteins. The results illustrate the power of this multiplex proteomics technology and illustrate how proteomics can be used to understand gene function.


Subject(s)
Electrophoresis, Gel, Two-Dimensional/methods , Proteome/chemistry , Animals , Cerebral Cortex/metabolism , Gene Expression Regulation , Hydrogen-Ion Concentration , Image Processing, Computer-Assisted , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Isoforms , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
13.
Brain ; 126(Pt 7): 1683-90, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12805119

ABSTRACT

Human African trypanosomiasis, also known as sleeping sickness, affects the CNS at the late stage of the disease. Untreated the disease is invariably fatal, and melarsoprol, the only available and effective treatment for CNS disease, is associated in up to 10% of cases with a severe post-treatment reactive encephalopathy (PTRE), which can itself cause death. We used a reproducible mouse model of the PTRE to investigate the pathogenesis and treatment of this condition. Mice infected with Trypanosoma brucei brucei and treated subcuratively with diminazene aceturate develop a severe meningoencephalitis that closely resembles PTRE. We previously reported that substance P plays an important role in PTRE. We investigated the effect of disrupting the gene encoding for the NK1 receptor in mice on the clinical and neuroinflammatory response in this model. After induction of PTRE, NK1-/- mice showed a significant reduction in clinical impairment compared with NK1+/+ mice, but the severity of the neuroinflammatory response was significantly greater in NK1-/- mice. To explore the mechanisms of this dissociated phenotype, we treated infected NK1-/- mice with antagonists to NK2 and NK3 receptors, either singly or in combination. While none of these antagonist treatments altered the clinical score, combined treatment with the NK2 and NK3 antagonists significantly reduced the neuroinflammatory grading score in the NK1-/- mice. Thus, the clinical and neuroinflammatory responses to parasite invasion can be mediated by different pathways, and, importantly, the neuroinflammatory response is altered by alternative tachykinin receptor usage. These findings could be exploited to develop novel anti-inflammatory therapies in Human African trypanosomiasis by modulating the NK1 receptor as well as the parasite.


Subject(s)
Meningoencephalitis/parasitology , Receptors, Neurokinin-1/physiology , Trypanosomiasis, African/complications , Animals , Diminazene , Disease Models, Animal , Hippocampus/pathology , Meningoencephalitis/pathology , Meningoencephalitis/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Neurokinin-1/genetics , Receptors, Neurokinin-2/antagonists & inhibitors , Receptors, Neurokinin-2/physiology , Receptors, Neurokinin-3/antagonists & inhibitors , Receptors, Neurokinin-3/physiology , Trypanosomiasis, African/pathology , Trypanosomiasis, African/physiopathology
14.
Eur J Neurosci ; 16(12): 2245-52, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12492418

ABSTRACT

Substance P and neurokinin-1 receptors (NK1) modulate the respiratory activity and are expressed early during development. We tested the hypothesis that NK1 receptors are involved in prenatal development of the respiratory network by comparing the resting respiratory activity and the respiratory response to hypoxia of control mice and mutant mice lacking the NK1 receptor (NK1-/-). In vitro and in vivo experiments were conducted on neonatal, young and adult mice from wild-type and NK1-/- strains. In the wild strain, immunohistological, pharmacological and electrophysiological studies showed that NK1 receptors were expressed within medullary respiratory areas prior to birth and that their activation at birth modulated central respiratory activity and the membrane properties of phrenic motoneurons. Both the membrane properties of phrenic motoneurons and the respiratory activity generated in vitro by brainstem-spinal cord preparation from NK1-/- neonate mice were similar to that from the wild strain. In addition, in vivo ventilation recordings by plethysmography did not reveal interstrain differences in resting breathing parameters. The facilitation of ventilation by short-lasting hypoxia was similar in wild and NK1-/- neonates but was significantly weaker in adult NK1-/- mice. Results demonstrate that NK1 receptors do appear to be necessary for a normal respiratory response to short-lasting hypoxia in the adult. However, NK1 receptors are not obligatory for the prenatal development of the respiratory network, for the production of the rhythm, or for the regulation of breathing by short-lasting hypoxia in neonates.


Subject(s)
Cell Differentiation/genetics , Medulla Oblongata/growth & development , Nerve Net/growth & development , Receptors, Neurokinin-1/deficiency , Respiratory Center/growth & development , Respiratory Physiological Phenomena/drug effects , Substance P/metabolism , Action Potentials/drug effects , Action Potentials/genetics , Animals , Animals, Newborn , Cell Differentiation/drug effects , Female , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/physiology , Hypoxia, Brain/genetics , Hypoxia, Brain/metabolism , Immunohistochemistry , Male , Medulla Oblongata/cytology , Medulla Oblongata/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Net/cytology , Nerve Net/metabolism , Phrenic Nerve/physiology , Receptors, Neurokinin-1/genetics , Respiratory Center/cytology , Respiratory Center/metabolism , Substance P/pharmacology , Synaptic Transmission/drug effects , Synaptic Transmission/genetics
15.
Eur J Neurosci ; 16(11): 2078-84, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12473075

ABSTRACT

Substance P has been proposed to be an important neurotransmitter in the carotid body with the neurokinin 1 (NK1) receptor, mediating excitation between the glomus cells and afferent nerve endings. In order to better understand the role of substance P, this study examined chemoreceptor afferent activity, in vitro, and tissue catecholamine levels and release in adult, wild-type mice and mice lacking the gene for the NK1 receptor (NK1-KO). Groups did not differ significantly in body weight, carotid body dopamine content or carotid body norepinephrine content. In wild-type mice, single unit activity increased from 0.59 +/- 0.14 Hz to 19.78 +/- 2.27 Hz during superfusion with strong hypoxia (PO2 approximately 25 Torr). Chemoreceptor activity in NK1-KO mice, increased from 0.71 +/- 0.23 to 21.50 +/- 3.62 Hz, and neither baseline or peak frequencies were significantly different from the wild-type group. Less severe hypoxia (PO2 approximately 45 torr), evoked peak activities of 12.50 +/- 1.88 and 10.64 +/- 3.72 Hz in wild-type and NK1-KO mice, which were also not significantly different. In response to severe hypoxia, free-tissue catecholamine increased to 4.92 +/- 0.85 microm in wild-type mice and 4.26 +/- 0.63 microm in NK1-KO mice, which were also not significantly different. It may therefore be concluded that loss of NK1 receptors has little effect on chemoreceptor function in the mouse, and thus they play, at best, a minor role in the hypoxic chemoreception process.


Subject(s)
Carotid Body/metabolism , Neurons, Afferent/metabolism , Receptors, Neurokinin-1/deficiency , Sensory Receptor Cells/metabolism , Substance P/metabolism , Visceral Afferents/metabolism , Animals , Carotid Body/cytology , Catecholamines/metabolism , Dopamine/metabolism , Female , Hypoxia/metabolism , Hypoxia/physiopathology , Mice , Mice, Knockout , Neurons, Afferent/cytology , Norepinephrine/metabolism , Receptors, Neurokinin-1/genetics , Sensory Receptor Cells/cytology , Signal Transduction/physiology , Superior Cervical Ganglion/cytology , Superior Cervical Ganglion/metabolism , Synaptic Transmission/physiology , Visceral Afferents/cytology
16.
Cancer Res ; 62(24): 7343-9, 2002 Dec 15.
Article in English | MEDLINE | ID: mdl-12499278

ABSTRACT

More than half of all chronic cancer pain arises from metastases to bone, and bone cancer pain is one of the most difficult of all persistent pain states to fully control. Several tumor types including sarcomas and breast, prostate, and lung carcinomas grow in or preferentially metastasize to the skeleton where they proliferate, and induce significant bone remodeling, bone destruction, and cancer pain. Many of these tumors express the isoenzyme cycloxygenase-2 (COX-2), which is involved in the synthesis of prostaglandins. To begin to define the role COX-2 plays in driving bone cancer pain, we used an in vivo model where murine osteolytic 2472 sarcoma cells were injected and confined to the intramedullary space of the femur in male C3HHeJ mice. After tumor implantation, mice develop ongoing and movement-evoked bone cancer pain-related behaviors, extensive tumor-induced bone resorption, infiltration of the marrow space by tumor cells, and stereotypic neurochemical alterations in the spinal cord reflective of a persistent pain state. Thus, after injection of tumor cells, bone destruction is first evident at day 6, and pain-related behaviors are maximal at day 14. A selective COX-2 inhibitor was administered either acutely [NS398; 100 mg/kg, i.p.] on day 14 or chronically in chow [MF. tricyclic; 0.015%, p.o.] from day 6 to day 14 after tumor implantation. Acute administration of a selective COX-2 inhibitor attenuated both ongoing and movement-evoked bone cancer pain, whereas chronic inhibition of COX-2 significantly reduced ongoing and movement-evoked pain behaviors, and reduced tumor burden, osteoclastogenesis, and bone destruction by >50%. The present results suggest that chronic administration of a COX-2 inhibitor blocks prostaglandin synthesis at multiple sites, and may have significant clinical utility in the management of bone cancer and bone cancer pain.


Subject(s)
Bone Neoplasms/complications , Bone Neoplasms/drug therapy , Cyclooxygenase Inhibitors/pharmacology , Isoenzymes/antagonists & inhibitors , Osteosarcoma/complications , Osteosarcoma/drug therapy , Pain/drug therapy , Animals , Bone Neoplasms/enzymology , Bone Neoplasms/pathology , Cell Division/drug effects , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Disease Models, Animal , Hyperostosis/drug therapy , Hyperostosis/enzymology , Hyperostosis/pathology , Male , Mice , Mice, Inbred C3H , Neurons, Afferent/drug effects , Neurons, Afferent/physiology , Osteoclasts/cytology , Osteoclasts/drug effects , Osteoclasts/pathology , Osteosarcoma/enzymology , Osteosarcoma/pathology , Pain/enzymology , Pain/etiology , Prostaglandin-Endoperoxide Synthases , Spinal Cord/drug effects , Spinal Cord/physiopathology
17.
J Biol Chem ; 277(43): 40342-51, 2002 Oct 25.
Article in English | MEDLINE | ID: mdl-12161429

ABSTRACT

The importance of voltage-activated calcium channels in pain processing has been suggested by the spinal antinociceptive action of blockers of N- and P/Q-type calcium channels as well as by gene targeting of the alpha1B subunit (N-type). The accessory beta3 subunits of calcium channels are preferentially associated with the alpha1B subunit in neurones. Here we show that deletion of the beta3 subunit by gene targeting affects strongly the pain processing of mutant mice. We pinpoint this defect in the pain-related behavior and ascending pain pathways of the spinal cord in vivo and at the level of calcium channel currents and proteins in single dorsal root ganglion neurones in vitro. The pain induced by chemical inflammation is preferentially damped by deletion of beta3 subunits, whereas responses to acute thermal and mechanical harmful stimuli are reduced moderately or not at all, respectively. The defect results in a weak wind-up of spinal cord activity during intense afferent nerve stimulation. The molecular mechanism responsible for the phenotype was traced to low expression of N-type calcium channels (alpha1B) and functional alterations of calcium channel currents in neurones projecting to the spinal cord.


Subject(s)
Calcium Channels/physiology , Ion Channel Gating , Pain/physiopathology , Sensory Thresholds , Amino Acid Sequence , Animals , Calcium Channels/chemistry , Calcium Channels/genetics , Evoked Potentials , Male , Mice , Neurons/physiology , Patch-Clamp Techniques , Spinal Cord/physiopathology
18.
Neurosci Lett ; 325(3): 199-202, 2002 Jun 14.
Article in English | MEDLINE | ID: mdl-12044655

ABSTRACT

Previous studies have shown that an abnormal salt-soluble form of G(1) acetylcholinesterase (AChE) is increased in the Alzheimer's disease (AD) brain. The aim of the present study was to examine changes in AChE activity in an in vivo model of beta-amyloid peptide (A beta) administration. Rats received intracerebroventricular injections of A beta(25-35) (20 microg/day for seven days). Levels of AChE were measured in cerebral cortex and cerebrospinal fluid (CSF) after two months. A beta(25-35) administration did not alter total AChE activity in the cerebral cortex or CSF. However, analysis of salt-extractable AChE isoforms revealed an increase in the proportion of G(1) in both cortex and CSF, similar to that previously observed in AD patients. The results support the view that changes in AChE isoform pattern in the AD brain are a direct consequence of A beta accumulation.


Subject(s)
Acetylcholinesterase/cerebrospinal fluid , Acetylcholinesterase/drug effects , Amyloid beta-Peptides/pharmacology , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Peptide Fragments/pharmacology , Acetylcholinesterase/metabolism , Amyloid beta-Peptides/administration & dosage , Animals , Injections, Intraventricular , Male , Peptide Fragments/administration & dosage , Rats , Rats, Wistar
19.
Proc Natl Acad Sci U S A ; 99(4): 2374-9, 2002 Feb 19.
Article in English | MEDLINE | ID: mdl-11854530

ABSTRACT

Vanilloid receptor subunit 1 (VR1) appears to play a critical role in the transduction of noxious chemical and thermal stimuli by sensory nerve endings in peripheral tissues. Thus, VR1 antagonists are useful compounds to unravel the contribution of this receptor to pain perception, as well as to induce analgesia. We have used a combinatorial approach to identify new, nonpeptidic channel blockers of VR1. Screening of a library of trimers of N-alkylglycines resulted in the identification of two molecules referred to as DD161515 [N-[2-(2-(N-methylpyrrolidinyl)ethyl]glycyl]-[N-[2,4-dichlorophenethyl]glycyl]-N-(2,4-dichlorophenethyl)glycinamide] and DD191515 [[N-[3-(N,N-diethylamino)propyl]glycyl]-[N-[2,4-dichlorophenethyl]glycyl]-N-(2,4-dichlorophenethyl)glycinamide] that selectively block VR1 channel activity with micromolar efficacy, rivaling that characteristic of vanilloid-related inhibitors. These compounds appear to be noncompetitive VR1 antagonists that recognize a receptor site distinct from that of capsaicin. Intraperitoneal administration of both trialkylglycines into mice significantly attenuated thermal nociception as measured in the hot plate test. It is noteworthy that these compounds eliminated pain and neurogenic inflammation evoked by intradermal injection of capsaicin into the animal hindpaw, as well as the thermal hyperalgesia induced by tissue irritation with nitrogen mustard. In contrast, responses to mechanical stimuli were not modified by either compound. Modulation of sensory nerve fibers excitability appears to underlie the peptoid analgesic activity. Collectively, these results indicate that blockade of VR1 activity attenuates chemical and thermal nociception and hyperalgesia, supporting the tenet that this ionotropic receptor contributes to chemical and thermal sensitivity and pain perception in vivo. These trialkylglycine-based, noncompetitive VR1 antagonists may likely be developed into analgesics to treat inflammatory pain.


Subject(s)
Hot Temperature , Hyperalgesia , Pain/drug therapy , Receptors, Drug/antagonists & inhibitors , Animals , Calcium/metabolism , Capsaicin/pharmacology , Dose-Response Relationship, Drug , Electrophysiology , Glycine/pharmacology , Inflammation/drug therapy , Knee/physiology , Male , Mice , Mice, Inbred ICR , Mustard Plant , Neurons/metabolism , Pain Threshold , Peptoids , Plant Extracts/pharmacology , Plant Oils , Rats , Rats, Wistar , Recombinant Proteins/metabolism , Stress, Mechanical , Time Factors , Xenopus
20.
Neuropharmacology ; 43(8): 1258-68, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12527475

ABSTRACT

Mice lacking the NK1 receptor, the preferred receptor for substance P, demonstrate normal analgesic responses to morphine on the hot plate assay, but have been predicted, on the basis of conditioned place preference studies, to be insensitive to the rewarding properties of opiates. In this study, self-administration and the development and maintenance of locomotor sensitisation of both morphine and cocaine were investigated in mice that lacked the NK1 gene (NK1 knockout mice, NK1(-/-)). Both wildtype and NK1(-/-) mice learned an operant lever-press response to obtain food. When intravenous infusions of morphine (0.2 mg/kg/infusion) were substituted for the food reward, the wildtype mice initially reduced rates of lever pressing, but then increased them on the rewarded lever to obtain approx. 10 infusions per 90 min session; in contrast, NK1(-/-) mice continued to operate both the rewarded, and non-rewarded levers at low rates. Additionally, NK1(-/-) mice failed, following repeated administration, to sensitise to the locomotor stimulant effects of morphine (15 mg/kg, i.p.). These deficits were specific to opiates, since NK1(-/-) mice responding for food or cocaine self-administration (0.65 mg/kg/infusion) did not differ from wildtypes, and they showed normal behavioural sensitisation to repeated cocaine administration (10 mg/kg, i.p.). These results demonstrate that NK1 receptors are critical for the reinforcing properties of morphine, and for adaptive responses elicited by repeated opiate administration. It is postulated that substance P and the NK1 receptor may be necessary for the development of opiate, but not cocaine addiction.


Subject(s)
Cocaine/administration & dosage , Cocaine/pharmacology , Morphine/administration & dosage , Motor Activity/drug effects , Receptors, Neurokinin-1/deficiency , Animals , Conditioning, Operant/drug effects , Conditioning, Operant/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/physiology , Receptors, Neurokinin-1/genetics , Self Administration/methods , Self Administration/psychology
SELECTION OF CITATIONS
SEARCH DETAIL
...