Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 186
Filter
Add more filters










Publication year range
1.
Andrologia ; 47(1): 25-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24387161

ABSTRACT

This study aimed to explore the effects of intracavernous injection (ICI) of P2X3 and NK1 receptor antagonists on erectile dysfunction (ED) induced by spinal cord transection in rats. Sixty male Sprague-Dawley (SD) rats were randomly divided into the following three groups (20 rats each group): sham operation group (C group), thoracic spinal cord transection group (T group) and sacral spinal cord transection group (S group). An ED model was established through complete transection of the thoracic or sacral spinal cord. Intracavernous pressure (ICP) with and without injection of P2X3 (Suramin) or NK1 (GR82334) receptor antagonists was recorded 3 weeks after surgery. Immunohistochemistry was employed to detect the expression of P2X3 and NK1 receptors in the dorsal root ganglion (DRG) and smooth muscle of corpus cavernosum. Data were processed with SPSS 17.0. ICI with Suramin (0.1, 0.3 and 1 mm) or GR82334 (0.1, 0.3 and 1 mm) increased ICP dose dependently in the T and S groups. The expression of P2X3 and NK1 receptors in DRG and smooth muscle of corpus cavernosum was up-regulated in the T and S groups. It is concluded that ICI of P2X3 and NK1 receptor antagonists may improve the recovery of erectile function in a rat model with ED after spinal cord transection.


Subject(s)
Erectile Dysfunction/etiology , Neurokinin-1 Receptor Antagonists/pharmacology , Penile Erection/drug effects , Penis/drug effects , Physalaemin/analogs & derivatives , Purinergic P2X Receptor Antagonists/pharmacology , Spinal Cord Injuries/complications , Suramin/pharmacology , Animals , Disease Models, Animal , Ganglia, Spinal/metabolism , Immunohistochemistry , Injections , Male , Penis/metabolism , Physalaemin/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Neurokinin-1/metabolism , Receptors, Purinergic P2X3/metabolism
2.
World J Urol ; 32(1): 91-7, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23666265

ABSTRACT

PURPOSE: The purpose of the study is to explore the function of P2X3 and NK1 receptors antagonists on cyclophosphamide (CYP)-induced cystitis in rats. METHODS: Sixty female Sprague-Dawley (SD) rats were randomly divided into three groups. The rats in the control group were intraperitoneally (i.p.) injected with 0.9% saline (4 ml/kg); the rats in the model group were i.p. injected with CYP (150 mg/kg); and the rats in the intervention group were i.p. injected with CYP with subsequently perfusion of bladder with P2X3 and NK1 receptors' antagonists, Suramin and GR 82334. Spontaneous pain behaviors following the administration of CYP were observed. Urodynamic parameters, bladder pressure-volume curve, maximum voiding pressure (MVP), and maximum cystometric capacity (MCC), were recorded. Pathological changes in bladder tissue were observed. Immunofluorescence was used to detect the expression of P2X3 and NK1 receptors in bladder. RESULTS: Cyclophosphamide treatment increased the spontaneous pain behaviors scores. The incidence of bladder instability during urine storage period of model group was significantly higher than intervention group (χ(2) = 7.619, P = 0.007) and control group (χ(2) = 13.755, P = 0.000). MCC in the model group was lower than the control and intervention groups (P < 0.01). Histological changes evident in model and intervention groups rats' bladder included edema, vasodilation, and infiltration of inflammatory cells. In model group, the expression of P2X3 receptor increased in urothelium and suburothelium, and NK1 receptor increased in suburothelium, while the expression of them in intervention group was lower. CONCLUSIONS: In CYP-induced cystitis, the expression of P2X3 and NK1 receptors increased in urothelium and/or suburothelium. Perfusion of bladder with P2X3 and NK1 receptors antagonists ameliorated the bladder function.


Subject(s)
Cyclophosphamide/adverse effects , Cystitis/chemically induced , Cystitis/drug therapy , Neurokinin-1 Receptor Antagonists/therapeutic use , Physalaemin/analogs & derivatives , Purinergic P2 Receptor Antagonists/therapeutic use , Suramin/therapeutic use , Animals , Cystitis/pathology , Disease Models, Animal , Female , Neurokinin-1 Receptor Antagonists/pharmacology , Pain/drug therapy , Physalaemin/pharmacology , Physalaemin/therapeutic use , Purinergic P2 Receptor Antagonists/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Neurokinin-1/drug effects , Receptors, Neurokinin-1/metabolism , Receptors, Purinergic P2X3/drug effects , Receptors, Purinergic P2X3/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Suramin/pharmacology , Urinary Bladder/drug effects , Urinary Bladder/metabolism , Urinary Bladder/pathology , Urination/drug effects , Urination/physiology , Urodynamics/drug effects , Urodynamics/physiology
3.
Cell Biochem Biophys ; 64(1): 29-44, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22628076

ABSTRACT

Protein aggregation and amyloid formation are associated with both pathological conditions in humans such as Alzheimer's disease and native functions such as peptide hormone storage in the pituitary secretory granules in mammals. Here, we studied amyloid fibrils formation by three neuropeptides namely physalaemin, kassinin and substance P of tachykinin family using biophysical techniques including circular dichroism, thioflavin T, congo red binding and microscopy. All these neuropeptides under study have significant sequence similarity with Aß(25-35) that is known to form neurotoxic amyloids. We found that all these peptides formed amyloid-like fibrils in vitro in the presence of heparin, and these amyloids were found to be nontoxic in neuronal cells. However, the extent of amyloid formation, structural transition, and morphology were different depending on the primary sequences of peptide. When Aß(25-35) and Aß40 were incubated with each of these neuropeptides in 1:1 ratio, a drastic increase in amyloid growths were observed compared to that of individual peptides suggesting that co-aggregation of Aß and these neuropeptides. The electron micrographs of these co-aggregates were dissimilar when compared with individual peptide fibrils further supporting the possible incorporation of these neuropeptides in Aß amyloid fibrils. Further, the fibrils of these neuropeptides can seed the fibrils formation of Aß40 and reduced the toxicity of preformed Aß fibrils. The present study of amyloid formation by tachykinin neuropeptides is not only providing an understanding of the mechanism of amyloid fibril formation in general, but also offering plausible explanation that why these neuropeptide might reduce the cytotoxicity associated with Alzheimer's disease related amyloids.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid/chemistry , Kassinin/chemistry , Peptide Fragments/chemistry , Physalaemin/chemistry , Algorithms , Amino Acid Sequence , Amyloid/pharmacology , Amyloid/ultrastructure , Amyloid beta-Peptides/ultrastructure , Benzothiazoles , Cell Line, Tumor , Circular Dichroism , Congo Red/chemistry , Heparin/chemistry , Humans , Kassinin/pharmacology , Microscopy, Electron , Neurons/chemistry , Neurons/drug effects , Peptide Fragments/ultrastructure , Physalaemin/pharmacology , Protein Binding , Protein Structure, Secondary , Substance P/chemistry , Substance P/pharmacology , Thiazoles/chemistry , Toxicity Tests/methods
4.
Eur J Pharmacol ; 668(1-2): 163-8, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21749865

ABSTRACT

Persistent pain associated with inflammatory arthritis is an aggravating factor that decreases patients' quality of life. Current therapies for joint pain have limited effectiveness and produce unwanted negative side effects. Although the involvement of substance P and its cognate tachykinin receptor, NK(1), in joint inflammation has been extensively documented through animal experiments, the development of oral tachykinin NK(1) receptor antagonists against arthritis-induced pain has been unsuccessful in humans to date. To explore the possibility of using tachykinin NK(1) receptor antagonists as local therapeutic agents for inflammatory arthritis, we examined the effects of tachykinin NK(1) receptor antagonists administered into the rat ankle joint on hyperalgesia in complete Freund's adjuvant (CFA)-induced inflammatory monoarthritis. Administration of the tachykinin NK(1) receptor antagonist WIN 51708 or GR 82334 into the affected ankle joint at day 3 following intra-articular CFA injection reduced the mechanical hyperalgesia 12 h after the tachykinin NK(1) receptor antagonist injection and their analgesic effects persisted for at least 2 days. Histological examinations revealed that intra-articular WIN 51708 reduced the CFA-induced destructive changes in the cartilage. These findings suggest that intra-articular injection of tachykinin NK(1) receptor antagonists is a promising strategy for relieving the hyperalgesia that occurs in inflammatory arthritis.


Subject(s)
Analgesics/pharmacology , Ankle Joint , Arthritis, Experimental/complications , Cartilage/drug effects , Hyperalgesia/complications , Hyperalgesia/drug therapy , Receptors, Tachykinin/antagonists & inhibitors , Analgesics/administration & dosage , Analgesics/therapeutic use , Androstanes/administration & dosage , Androstanes/pharmacology , Androstanes/therapeutic use , Animals , Benzimidazoles/administration & dosage , Benzimidazoles/pharmacology , Benzimidazoles/therapeutic use , Cartilage/pathology , Freund's Adjuvant/adverse effects , Indomethacin/administration & dosage , Indomethacin/pharmacology , Indomethacin/therapeutic use , Inflammation/complications , Inflammation/drug therapy , Inflammation/pathology , Injections, Subcutaneous , Male , Physalaemin/administration & dosage , Physalaemin/analogs & derivatives , Physalaemin/pharmacology , Physalaemin/therapeutic use , Rats , Rats, Sprague-Dawley
5.
J Dent Res ; 89(10): 1154-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20651096

ABSTRACT

UNLABELLED: Peripheral inflammation produces pain hypersensitivity by sensitizing nociceptors. Potentiation of P2X3 receptor activity in nociceptors may play an important role in this peripheral sensitization. However, we do not fully understand how P2X3 activity is elevated in inflammation. Thus, we investigated whether P2X3 activity in trigeminal nociceptive neurons is regulated by the neurokinin-1 (NK-1) receptor that is activated by an inflammatory mediator, substance P. Single-cell RT-PCR and immunohistochemistry revealed that NK-1 in nociceptive neurons was mainly co-expressed with P2X3. Ca(2+) imaging and whole-cell patch-clamp recordings indicated that both substance P and Sar-substance P, a selective NK-1 agonist, significantly potentiated α,ß-meATP-induced currents and [Ca(2+)](i) responses in nociceptive neurons. These potentiating effects were completely blocked by GR82334, a specific NK-1 antagonist. Our results demonstrate that substance P sensitizes P2X3 receptor through the activation of NK-1, thus warranting these receptors as possible targets for pain therapy in the orofacial region. ABBREVIATIONS: α,ß-methylene adenosine 5'-triphosphate (ATP), α,ß-meATP; neurokinin-1, NK-1; single-cell reverse-transcription polymerase chain-reaction, single-cell RT-PCR; [Sar(9),Met(O(2))(11)]-substance P, Sar-substance P.


Subject(s)
Neurons/drug effects , Nociceptors/drug effects , Receptors, Purinergic P2/drug effects , Substance P/pharmacology , Trigeminal Nerve/drug effects , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/metabolism , Animals , Animals, Newborn , Calcium Signaling/drug effects , Cytophotometry , Immunohistochemistry , Inflammation Mediators/pharmacology , Neurokinin-1 Receptor Antagonists , Patch-Clamp Techniques , Physalaemin/analogs & derivatives , Physalaemin/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Neurokinin-1/agonists , Receptors, Neurokinin-1/drug effects , Receptors, Purinergic P2X3 , Reverse Transcriptase Polymerase Chain Reaction , Substance P/analogs & derivatives
6.
Eur J Pharmacol ; 628(1-3): 202-6, 2010 Feb 25.
Article in English | MEDLINE | ID: mdl-19958761

ABSTRACT

We examined the effects of physalaemin, an agonist of tachykinin receptors, on mechanical responses in the rat esophagus to clarify possible regulatory roles of tachykinins in esophageal motility. Exogenous application of physalaemin caused tonic contractions in rat esophageal segments when tension was recorded in the longitudinal direction but not when tension was recorded in the circular direction. The physalaemin-evoked contractions were blocked by pretreatment with nifedipine, a blocker of L-type calcium channels in both striated and smooth muscle cells. However, tetrodotoxin, a blocker of voltage-dependent sodium channels in striated muscle cells and neurons, did not affect the physalaemin-induced contractions. These results indicate that physalaemin might induce contractile responses in longitudinal smooth muscle of the muscularis mucosa via direct actions on muscle cells but not on neurons. Although pretreatment with a tachykinin NK(1) receptor antagonist, N-acetyl-l-tryptophan 3,5-bis (trifluoromethyl) benzyl ester (L-732,138), did not significantly affect the physalaemin-evoked contractions in rat esophageal segments, a tachykinin NK(2) receptor antagonist, (S)-N-methyl-N[4-(4-acetylamino-4-phenylpiperidino)-2-(3,4-dichlorophenyl) butyl] benzamide (SR48968), and a tachykinin NK(3) receptor antagonist, (S)-(N)-(1-(3-(1-benzoyl-3-(3,4-dichlorophenyl) piperidin-3-yl)propyl)-4-phenylpiperidin-4-yl)-N-methylacetamide (SR142801), significantly inhibited the physalaemin-evoked contractions. These results suggest that tachykinins can activate longitudinal contraction of smooth muscle in the muscularis mucosa, mediated via tachykinin NK(2) and NK(3) receptors on muscle cells, in the rat esophagus.


Subject(s)
Esophagus/drug effects , Esophagus/physiology , Muscle Contraction/drug effects , Physalaemin/pharmacology , Substance P/analogs & derivatives , Animals , Atropine/pharmacology , Esophagus/metabolism , Male , Mucous Membrane/drug effects , Mucous Membrane/metabolism , Mucous Membrane/physiology , Muscle Tonus/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Muscle, Smooth/physiology , Muscle, Striated/drug effects , Muscle, Striated/metabolism , Muscle, Striated/physiology , Nifedipine/pharmacology , Physalaemin/analogs & derivatives , Rats , Rats, Wistar , Receptors, Tachykinin/agonists , Receptors, Tachykinin/antagonists & inhibitors , Receptors, Tachykinin/metabolism , Tetrodotoxin/pharmacology
7.
FEBS Lett ; 583(12): 2148-53, 2009 Jun 18.
Article in English | MEDLINE | ID: mdl-19446555

ABSTRACT

The effects of the sensory neurotransmitter substance P on the expression of tight junction proteins and on barrier function in human corneal epithelial cells were investigated. The expression of ZO-1, but not that of occludin or claudin-1, was increased by substance P in a concentration- and time-dependent manner. This effect was inhibited by the NK-1 receptor antagonist GR82334 and by KN62, an inhibitor of Ca(2+)- and calmodulin-dependent protein kinase II. Substance P also increased the transepithelial electrical resistance of a cell monolayer in a manner sensitive to GR82334. Substance P may therefore play a role in maintenance of tight junctions in the corneal epithelium.


Subject(s)
Epithelium, Corneal/drug effects , Epithelium, Corneal/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Substance P/pharmacology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Base Sequence , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Cells, Cultured , Claudin-1 , DNA Primers/genetics , Electric Impedance , Epithelium, Corneal/cytology , Humans , Neurokinin-1 Receptor Antagonists , Occludin , Physalaemin/analogs & derivatives , Physalaemin/pharmacology , Substance P/metabolism , Tight Junctions/drug effects , Tight Junctions/metabolism , Up-Regulation/drug effects , Zonula Occludens-1 Protein
8.
Neuropsychopharmacology ; 34(2): 331-40, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18418359

ABSTRACT

The neural pathways through which substance P (SP) influences fear and anxiety are poorly understood. However, the amygdala, a brain area repeatedly implicated in fear and anxiety processes, is known to contain large numbers of SP-containing neurons and SP receptors. Several studies have implicated SP neurotransmission within the amygdala in anxiety processes. In the present study, we evaluated the effects of site-specific infusions of an SP receptor antagonist, GR 82334, on conditioned fear responses using the fear-potentiated startle paradigm. GR 82334 infusion into the basolateral (BLA) or the medial (MeA) nuclei of the amygdala, but not into the central nucleus of the amygdala (CeA), dose dependently reduced fear-potentiated startle. Similar effects were obtained with GR 82334 infusion into the ventromedial nucleus of the hypothalamus (VMH), to which the MeA projects, and into the rostral dorsolateral periaqueductal gray (PAG), to which the VMH projects, but not into the deep layers of the superior colliculus/deep mesencephalic nucleus (dSC/DpMe), an output of the CeA previously shown to be important for fear-potentiated startle. Consistent with previous findings, infusion of the AMPA receptor antagonist, NBQX, into the dSC/DpMe, but not into the PAG, did disrupt fear-potentiated startle. These findings suggest that multiple outputs from the amygdala play a critical role in fear-potentiated startle and that SP plays a critical, probably modulatory role, in the MeA to VMH to PAG to the startle pathway based on these and data from others.


Subject(s)
Amygdala/physiology , Fear , Periaqueductal Gray/drug effects , Reflex, Startle , Substance P/physiology , Ventromedial Hypothalamic Nucleus/physiology , Amygdala/drug effects , Analysis of Variance , Animals , Catheterization , Conditioning, Classical/drug effects , Fear/drug effects , Male , Neurokinin-1 Receptor Antagonists , Physalaemin/analogs & derivatives , Physalaemin/pharmacology , Quinoxalines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, AMPA/antagonists & inhibitors , Reflex, Startle/drug effects , Superior Colliculi/drug effects , Ventromedial Hypothalamic Nucleus/drug effects
9.
Am J Respir Crit Care Med ; 175(4): 316-22, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17110645

ABSTRACT

RATIONALE: Despite accumulating evidence that psychological stress has a short-lasting detrimental effect on asthma, little is known about the way stress in childhood predisposes to adult asthma. OBJECTIVES: Using a communication box, we investigated the long-lasting effect of early psychological and physical stress on adult asthma in mice. METHODS: Male BALB/c mice were exposed to either psychological stress or physical stress three times (every other day) during their fourth week of life. The mice were sensitized to ovalbumin at 8 and 10 weeks, and an ovalbumin airway challenge was conducted at the age of 11 weeks. RESULTS: Twenty-four hours after ovalbumin challenge, both psychological and physical stress-exposed mice exhibited a significant acceleration in the number of total mononuclear cells and eosinophils and airway hyperresponsiveness compared with control mice. No differences in serum anti-OVA-specific immunoglobulin E levels were found between stress-exposed and control animals after antigen sensitization. In the psychological stress group, but not in the physical stress group, an elevation of the serum corticosterone levels during ovalbumin challenge was significantly attenuated in comparison with the control group. Moreover, pretreatment with RU-486, a glucocorticoid receptor antagonist, before ovalbumin challenge completely inhibited a psychological stress-induced exacerbation of asthma. However, pretreatment with GR-82334, a neurokinin-1 receptor antagonist, failed to affect physical stress-induced augmentation of airway inflammation. CONCLUSION: Early psychological and physical stresses aggravated adult asthma via hyporesponsiveness of the hypothalamic-pituitary-adrenal axis during antigen challenge and via a pathway(s) distinct from the hypothalamic-pituitary-adrenal axis or neurokinin-1 receptors.


Subject(s)
Asthma/physiopathology , Hypothalamo-Hypophyseal System/physiopathology , Pituitary-Adrenal System/physiopathology , Stress, Physiological/physiopathology , Stress, Psychological/physiopathology , Animals , Asthma/metabolism , Asthma/prevention & control , Bronchial Provocation Tests , Bronchoalveolar Lavage Fluid , Corticosterone/blood , Eosinophils/metabolism , Hormone Antagonists/pharmacology , Leukocytes, Mononuclear/metabolism , Male , Mice , Mice, Inbred BALB C , Mifepristone/pharmacology , Neurokinin-1 Receptor Antagonists , Neurotransmitter Agents/pharmacology , Ovalbumin/administration & dosage , Ovalbumin/immunology , Physalaemin/analogs & derivatives , Physalaemin/pharmacology , Receptors, Glucocorticoid/antagonists & inhibitors
10.
Brain Res ; 1112(1): 106-13, 2006 Sep 27.
Article in English | MEDLINE | ID: mdl-16928362

ABSTRACT

Previous studies have indicated that the geniohyoid (GH) muscle receives innervation via both the hypoglossal nerve (CNXII) and the ansa cervicalis. Our recent studies revealed that the efferent root that contributes to the ansa cervicalis is a parasympathetic pathway and contains postganglionic cell bodies. Afferent axons from the GH muscle also travel via the ansa cervicalis, and afferent cell bodies are located in spinal ganglia. The present study attempts to locate the central terminations of these afferents. From the peripheral cut end of the ansa cervicalis, we recorded afferent discharges that coincided with inspiration and these were elicited by stretch of the GH muscle. After cutting CNXII proximal to its union with the ansa cervicalis, we applied horseradish peroxidase to the branch of CNXII that innervates the GH muscle. This procedure labeled cells ipsilaterally in the C2 spinal ganglia but not in the brainstem or upper spinal cord. Substance P-reactive terminals in the peripheral CNXII trunk were in apparent contact with vasoactive intestinal peptide-reactive cell bodies. Addition of the NK1 receptor agonist SP(NK1) excited parasympathetic postganglionic neurons and the specific NK1 receptor antagonist GR82334 blocked these effects in vitro. These results suggest that GH primary afferents synapse on parasympathetic postganglionic neurons in the CNXII trunk and that activation of SP(NK1) receptors modulates activity in these neurons.


Subject(s)
Afferent Pathways/physiology , Hypoglossal Nerve/physiology , Neurons, Afferent/physiology , Parasympathetic Fibers, Postganglionic/cytology , Receptors, Neurokinin-1/metabolism , Action Potentials/physiology , Animals , Dose-Response Relationship, Drug , Electromyography/methods , Hypoglossal Nerve/drug effects , Hypoglossal Nerve/metabolism , Immunohistochemistry/methods , In Vitro Techniques , Models, Biological , Neck Muscles/physiology , Neurokinin-1 Receptor Antagonists , Neurons, Afferent/drug effects , Peptide Fragments/pharmacology , Physalaemin/analogs & derivatives , Physalaemin/pharmacology , Rats , Rats, Wistar , Receptors, Neurokinin-1/agonists , Substance P/analogs & derivatives , Substance P/metabolism , Substance P/pharmacology , Vasoactive Intestinal Peptide/metabolism , Wheat Germ Agglutinin-Horseradish Peroxidase Conjugate/metabolism
11.
Pharmacol Biochem Behav ; 84(1): 94-101, 2006 May.
Article in English | MEDLINE | ID: mdl-16757018

ABSTRACT

The neuropeptide substance P (SP) and its preferred receptor, the neurokinin-1 (NK-1) receptor, have been implicated in some of the reward-related behavioural effects of abused drugs, including psychostimulants and opiates. The first objective of the present series of experiments was to assess the role of the NK-1 receptor in two reward-related behavioural effects of cocaine: locomotor activity and self-administration. In tests for locomotor activity, rats were given intracerebroventricular (ICV) infusions of the selective NK-1 receptor antagonist, GR82334 (0, 10, 50 pmol), prior to systemic injections of cocaine. In self-administration experiments, rats were trained to self-administer cocaine on a fixed-ratio 5 (FR5) schedule of reinforcement. Following acquisition of stable responding, animals were pretreated with GR82334 (0, 2, 10, 50 pmol; ICV) prior to subsequent self-administration sessions. Based on evidence suggesting a potentially selective role for NK-1 receptors in opiate reward, we also examined the effects of GR82334 on morphine-induced locomotor activity and heroin self-administration. Results showed that GR82334 had no effect on cocaine-induced locomotor activity or cocaine self-administration, but attenuated morphine-induced locomotor activity and increased heroin self-administration. These findings suggest that endogenous activity at NK-1 receptors may play a specific role in opiate-induced, but not cocaine-induced, locomotor activation and reinforcement.


Subject(s)
Cocaine/pharmacology , Locomotion/drug effects , Narcotics/pharmacology , Neurokinin-1 Receptor Antagonists , Physalaemin/analogs & derivatives , Animals , Injections, Intraventricular , Male , Physalaemin/administration & dosage , Physalaemin/pharmacology , Rats , Rats, Wistar , Self Administration
12.
Brain Res ; 1021(1): 101-11, 2004 Sep 17.
Article in English | MEDLINE | ID: mdl-15328037

ABSTRACT

The major local symptom of Phoneutria nigriventer envenomation is an intense pain, which can be controlled by infiltration with local anesthetics or by systemic treatment with opioid analgesics. Previous work showed that intraplantar (i.pl) injection of Phoneutria nigriventer venom in rats induces hyperalgesia, mediated peripherally by tachykinin and glutamate receptors. The present study examined the spinal mechanisms involved in pain-enhancing effect of this venom. Intraplantar injection of venom into rat hind paw induced hyperalgesia. This phenomenon was inhibited by intrathecal (i.t.) injection of tachykinin NK1 (GR 82334) or NK2 (GR 94800) receptor antagonists, a calcitonin gene-related peptide (CGRP) receptor antagonist (CGRP8-37) and N-methyl-D-aspartate (NMDA; MK 801 and AP-5), non-NMDA ionotropic (CNQX), or metabotropic (AIDA and MPEP) glutamate receptor antagonists, suggesting the involvement of spinal neurokinins and excitatory amino acids. The role of proinflammatory cytokines, nitric oxide (NO), and prostanoids in spinally mediated pain facilitation was also investigated. Pharmacological blockade of tumour necrosis factor-alpha (TNFalpha) or interleukin-1beta (IL-1beta) reduced the hyperalgesic response to venom. Intrathecal injection of L-N6-(1-iminoethyl)lysine (L-NIL), but not of 7-nitroindazole (7-NI), inhibited hyperalgesia induced by the venom, indicating that NO, generated by the activity of the inducible form of nitric oxide synthase, also mediates this phenomenon. Furthermore, indomethacin, an inhibitor of cyclooxigenases (COX), or celecoxib, a selective inhibitor of COX-2, abolished venom-induced hyperalgesia, suggesting the involvement of spinal prostanoids in this effect. These data indicate that the spinal mechanisms of pain facilitation induced by Phoneutria nigriventer venom involves a plethora of mediators that may cooperate in the genesis of venom-induced central sensitization.


Subject(s)
Hyperalgesia/immunology , Interleukin-1/immunology , Lysine/analogs & derivatives , Pain/immunology , Physalaemin/analogs & derivatives , Spider Venoms/toxicity , Spinal Cord/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Antibodies/pharmacology , Calcitonin Gene-Related Peptide Receptor Antagonists , Celecoxib , Citrates/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Dizocilpine Maleate/pharmacology , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Indazoles/pharmacology , Interleukin-1/antagonists & inhibitors , Lysine/pharmacology , Male , Neurokinin-1 Receptor Antagonists , Nitric Oxide Synthase/antagonists & inhibitors , Pain/chemically induced , Pain/drug therapy , Physalaemin/pharmacology , Prostaglandins/metabolism , Pyrazoles , Rats , Rats, Wistar , Receptors, Calcitonin Gene-Related Peptide/metabolism , Receptors, Neurokinin-1/metabolism , Spinal Cord/metabolism , Sulfonamides/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors
13.
Naunyn Schmiedebergs Arch Pharmacol ; 370(1): 35-45, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15235762

ABSTRACT

Two tachykinin peptides, bufokinin and Xenopus neurokinin A (X-NKA) were recently isolated from Xenopus laevis. In this study we investigated the tachykinin receptors in the Xenopus gastrointestinal tract. In functional studies using stomach circular muscle strips, all peptides had similar potencies (EC50 values 1-7 nM). The rank order of potency to contract the intestine was physalaemin (EC50 1 nM)> or =bufokinin (EC50 3 nM)>substance P (SP)> or =cod SP>NKA>>X-NKA (EC50 1,900 nM). No maximum response could be obtained for [Sar9,Met(O2)11]SP, eledoisin and kassinin. In stomach strips, the mammalian tachykinin receptor antagonists RP 67580 (NK1) and MEN 10376 (NK2) had agonistic effects but did not antagonize bufokinin or X-NKA. In intestinal strips, RP 67580 (1 microM) reduced the maximal response to X-NKA but not bufokinin, while MEN 10376 was ineffective. [125I]BH-bufokinin bound with high affinity to a single class of sites, of KD 213+/-35 (stomach) and 172+/-9.3 pM (intestine). Specific binding of [125I]BH-bufokinin was displaced by bufokinin> or =SP>NKA> or =eledoisin approximately kassinin>X-NKA, indicating binding to a tachykinin NK1-like receptor. Selective tachykinin receptor antagonists were weak or ineffective. Other iodinated tachykinins ([125I]NKA and [125I]BH-eledoisin) displayed biphasic competition profiles, with the majority of sites preferring bufokinin rather than X-NKA. In conclusion, there is evidence for two different tachykinin receptors in Xenopus gastrointestinal tract. Both receptors may exist in stomach, whereas the bufokinin-preferring NK1-like receptor predominates in longitudinal muscle of the small intestine. Antagonists appear to interact differently with amphibian receptors, compared with mammalian receptors.


Subject(s)
Neurokinin A/analogs & derivatives , Physalaemin/analogs & derivatives , Receptors, Tachykinin/chemistry , Receptors, Tachykinin/drug effects , Species Specificity , Substance P/analogs & derivatives , Xenopus/metabolism , Animals , Binding Sites/drug effects , Cardia/cytology , Cardia/drug effects , Cardia/metabolism , Dose-Response Relationship, Drug , Eledoisin/pharmacology , Female , Indoles/pharmacology , Intercellular Signaling Peptides and Proteins/chemistry , Intercellular Signaling Peptides and Proteins/pharmacology , Intestine, Small/cytology , Intestine, Small/drug effects , Intestine, Small/metabolism , Iodine Radioisotopes , Isoindoles , Kassinin/pharmacology , Male , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , Neurokinin A/antagonists & inhibitors , Neurokinin A/chemistry , Neurokinin A/pharmacology , Peptide Fragments/pharmacology , Physalaemin/pharmacology , Receptors, Tachykinin/physiology , Substance P/pharmacology
14.
Peptides ; 24(4): 543-51, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12860198

ABSTRACT

In this study, we examined the activity of the tachykinins (TKs) on lamb and sheep isolated gallbladder and whether the TKs are involved in the capsaicin-induced activity in these tissues. Substance P (SP) and physalaemin (PHYS) contracted lamb gallbladder, PHYS-induced striking tachyphylaxis. This tissue was nearly insensitive to neurokinin A (NKA), neurokinin B (NKB), septide, and capsaicin. As in lamb tissues, SP and PHYS both contracted sheep gallbladder although PHYS induced no tachyphylaxis. At doses that had no effect on lamb tissue, NKA, NKB, septide, and capsaicin contracted sheep gallbladder. Our findings indicate that TK receptors differ in adult and young ovine gallbladder. The activity of PHYS on lamb gallbladder could depend on the existence of an unusual binding site, carrying one or more residues critical for the N-terminal sequence present in PHYS but not in SP.


Subject(s)
Gallbladder/drug effects , Substance P/analogs & derivatives , Tachykinins/pharmacology , Animals , Binding Sites , Capsaicin/metabolism , Capsaicin/pharmacology , Dose-Response Relationship, Drug , Female , Male , Muscles/metabolism , Neurokinin A/pharmacology , Neurokinin B/pharmacology , Peptide Fragments/pharmacology , Physalaemin/pharmacology , Protein Structure, Tertiary , Pyrrolidonecarboxylic Acid/analogs & derivatives , Receptors, Tachykinin/biosynthesis , Sheep , Substance P/pharmacology
15.
Br J Pharmacol ; 138(7): 1233-43, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12711623

ABSTRACT

1. Neurokinins contribute to the neural regulation of gastrointestinal (GI) smooth muscles. We studied responses of murine colonic smooth muscle cells to substance P (SP) and NK(1) and NK(2) agonists using confocal microscopy and the patch clamp technique. 2. Colonic myocytes generated localized Ca(2+) transients that were coupled to spontaneous transient outward currents (STOCs). SP (10(-10) M) increased Ca(2+) transients and STOCs. Higher concentrations of SP (10(-6) M) increased basal Ca(2+) and inhibited Ca(2+) transients and STOCs. 3. Effects of SP were due to increased Ca(2+) entry via L-type Ca(2+) channels, and were mediated by protein kinase C (PKC). Nifedipine (10(-6) M) and the PKC inhibitor, GF 109203X (10(-6) M) reduced L-type Ca(2+) current and blocked the effects of SP. 4. SP responses depended upon parallel stimulation of NK(1) and NK(2) receptors. NK(1) agonist ([Sar(9),Met(O(2))(11)]-substance P; SSP) and NK(2) agonists (neurokinin A (NKA) or GR-64349) did not mimic the effects of SP alone, but NK(1) and NK(2) agonists were effective when added in combination (10(-10)-10(-6) M). Consistent with this, either an NK(1)-specific antagonist (GR-82334; 10(-7) M) or an NK(2)-specific antagonist (MEN 10,627; 10(-7) M) blocked responses to SP (10(-6) M). 5. Ryanodine (10(-5) M) blocked the increase in Ca(2+) transients and STOCs in response to SP (10(-10) M). 6. Our findings show that low concentrations of SP, via PKC-dependent enhancement of L-type Ca(2+) current and recruitment of ryanodine receptors, stimulate Ca(2+) transients. At higher concentrations of SP (10(-6) M), basal Ca(2+) increases and spontaneous Ca(2+) transients and STOCs are inhibited.


Subject(s)
Calcium Signaling/drug effects , Colon/cytology , Colon/drug effects , Electric Conductivity , Neurokinin A/analogs & derivatives , Physalaemin/analogs & derivatives , Substance P/pharmacology , Animals , Calcium Signaling/physiology , Colon/metabolism , Imidazoles/pharmacology , Indoles/pharmacology , Male , Maleimides/pharmacology , Membrane Potentials/drug effects , Mice , Mice, Inbred BALB C , Microscopy, Confocal , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Neurokinin A/pharmacology , Nicardipine/pharmacology , Patch-Clamp Techniques , Peptide Fragments/pharmacology , Peptides/pharmacology , Physalaemin/pharmacology , Receptors, Tachykinin/drug effects , Receptors, Tachykinin/physiology , Ryanodine/pharmacology , Ryanodine Receptor Calcium Release Channel/drug effects , Second Messenger Systems/drug effects , Second Messenger Systems/physiology , Substance P/analogs & derivatives
16.
Brain Res ; 944(1-2): 19-31, 2002 Jul 19.
Article in English | MEDLINE | ID: mdl-12106662

ABSTRACT

Substance P (SP) is associated with metabo- and mechanoreceptor afferent fibers ('ergoreceptors') in skeletal muscle as well as the afferent fibers from carotid sinus baroreceptors. Afferent activity from each of these are at least partially integrated in the nucleus tractus solitarius (NTS). The purpose of this study was to determine whether SP was released from the NTS during acute reflex-induced changes in blood pressure caused by stimulating these receptors. Both the muscle pressor response and the baroreflex were studied in adult cats anaesthetized with alpha-chloralose. SP antibody-coated microprobes were used to measure the possible release of SP from the NTS. The muscle pressor response caused a release of immunoreactive SP-like substances (irSP) from the rostral medial NTS, as well as the dorsal motor nucleus (DMV) and lateral tegmental field (FTL). This release was not dependent on intact afferent input from the carotid sinus nerve, but was a function of activation of muscle ergoreceptors, since no irSP was released in response to stimulation of the motor nerves after the muscle was paralyzed. There was no detectable release of irSP from the mNTS during carotid artery occlusions (baroreceptor unloading). Baroreceptor activation, induced by the i.v. injection of the vasoconstrictor, phenylephrine, did not cause the release of irSP from the mNTS above resting baseline levels. These data suggest that SP is involved with the mediation of the afferent signal from muscle ergoreceptor fibers in the medial NTS. SP is not involved with the mediation of baroreceptor afferent signaling in the medial NTS. The release of SP in response to ergoreceptors activation may function to excite an inhibitory pathway which inhibits baroreflex signals that would tend to reduce the blood pressure and heart rate during the muscle pressor response.


Subject(s)
Afferent Pathways/metabolism , Mechanoreceptors/metabolism , Muscle, Skeletal/innervation , Neurons/metabolism , Physalaemin/analogs & derivatives , Pressoreceptors/metabolism , Solitary Nucleus/metabolism , Substance P/metabolism , Afferent Pathways/drug effects , Animals , Baroreflex/drug effects , Baroreflex/physiology , Cardiovascular Physiological Phenomena/drug effects , Cats , Mechanoreceptors/drug effects , Movement/drug effects , Movement/physiology , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle Fatigue/drug effects , Muscle Fatigue/physiology , Muscle, Skeletal/physiology , Neural Inhibition/drug effects , Neural Inhibition/physiology , Neurokinin-1 Receptor Antagonists , Neurons/cytology , Neurons/drug effects , Physalaemin/pharmacology , Pressoreceptors/drug effects , Receptors, Neurokinin-1/metabolism , Solitary Nucleus/cytology , Solitary Nucleus/drug effects , Sympathetic Nervous System/cytology , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/metabolism
17.
Acta Pharmacol Sin ; 23(4): 289-95, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11931700

ABSTRACT

AIM: To examine the effect of tachykinins on the ascending reflex pathway in rat small intestine, we used different selective neurokinin (NK) receptor antagonists (RA): a) NK1-RA: GR-82334 and CP-96.345, b) NK2-RA: MEN-10.376 and L-659.877. The aim was further to investigate the effect of substance P (SP) on the ascending excitatory and descending inhibitory reflex pathway. METHODS: The whole segments of rat ileum (10 cm in length) were studied in an organ bath. Ascending contraction of circle muscle was elicited by anal electrical stimulation (3 Hz, 1 ms, 20 V) and measured as change of intraluminal pressure by a perfused manometric system 2 cm and 4 cm orad of the stimulation site. RESULTS: GR-82334 and CP-96.345 (NK1-RA) caused a significant dose-related inhibition of the oral contraction at a distance of 4 cm: GR-82334 [area: -10 % +/- 8 % (10 nmol/L); -29 % +/ -10 % (1000 nmol/L). P < 0.05, n = 10], CP-96.345 [area: -2 %+/- 6 %(0.1 nmol/L); -14 % +/- 10 % (10 nmol/L). P < 0.01, n = 8], whereas the contractile response at a distance of 2 cm was unaltered (n = 8). In contrast, MEN-10.376 and L-659.877 (NK2-RA) did not alter the amplitude or the area under the curve (n = 10). Neither the NK1- nor the NK2-receptor antagonists had a significant effect on the latency of the reflex response. SP showed a significant increase in the ascending contraction and the descending relaxation (n = 6, P < 0.01). CONCLUSION: These results demonstrate that blockade of NK1-receptors decreases the oral reflex response. Latency of the reflex response remains unchanged, indicating that the effect is not due to an action on interneurons. NK2-receptors do not take part in the ascending reflex in rat small intestine. SP increases the descending relaxant reflex response and ascending contraction.


Subject(s)
Ileum/drug effects , Neurokinin A/analogs & derivatives , Physalaemin/analogs & derivatives , Reflex/drug effects , Substance P/pharmacology , Tachykinins/pharmacology , Afferent Pathways/drug effects , Animals , Biphenyl Compounds/pharmacology , Efferent Pathways/drug effects , Ileum/physiology , In Vitro Techniques , Male , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , Neurokinin A/pharmacology , Neurokinin-1 Receptor Antagonists , Peptide Fragments/pharmacology , Physalaemin/pharmacology , Rats , Rats, Wistar , Reaction Time/drug effects , Receptors, Neurokinin-2/antagonists & inhibitors , Substance P/antagonists & inhibitors
18.
Jpn J Pharmacol ; 86(3): 336-41, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11488435

ABSTRACT

The intrathecal injection of fenvalerate, a sodium channel activator, at doses of 0.01 to 3 microg, dose-dependently induced the duration of a characteristic behavioral syndrome mainly consisting of reciprocal hind limb scratching directed towards caudal parts of the body and biting or licking of the hind legs in mice. Fenvalerate-induced behavior was inhibited by morphine (1-10 mg/kg, i.p.). The characteristic behavior was also inhibited by mexiletine, a sodium channel blocker; MK-801, a N-methyl-D-aspartate ion-channel blocker; and GR82334, a neurokinin-1-receptor antagonist. Calphostin C (3 pmol, i.t.), a protein kinase C inhibitor, inhibited fenvalerate-induced behavior. On the other hand, phorbol-12, 13-dibutyrate (50 pmol, i.t.), a protein kinase C activator, markedly enhanced the fenvalerate-induced behavior. The present results also showed that fenvalerate produced thermal allodynia and hyperalgesia in the tail-flick test. Furthermore, fenvalerate-induced thermal allodynia and hyperalgesia were inhibited by the pretreatment with calphostin C. These results suggest that the intrathecal administration of fenvalerate induces a marked nociceptive response and thermal allodynia/hyperalgesia, and they suggest that tetrodotoxin-resistant sodium channels may play an important role in this effect.


Subject(s)
Hyperalgesia/chemically induced , Pain/chemically induced , Physalaemin/analogs & derivatives , Pyrethrins/pharmacology , Animals , Behavior, Animal/drug effects , Dizocilpine Maleate/pharmacology , Enzyme Activators/pharmacology , Enzyme Inhibitors/pharmacology , Injections, Spinal , Male , Mexiletine/pharmacology , Mice , Mice, Inbred ICR , Nitriles , Physalaemin/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Pyrethrins/administration & dosage
19.
J Physiol ; 534(Pt. 2): 447-64, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11454963

ABSTRACT

1. The effects of substance P (SP), acting at NK1 receptors, on the excitability and inspiratory activity of hypoglossal (XII) motoneurons (MNs) were investigated using rhythmically active medullary-slice preparations from neonatal mice (postnatal day 0-3). 2. Local application of the NK1 agonist [SAR(9),Met (O(2))(11)]-SP (SP(NK1)) produced a dose-dependent, spantide- (a non-specific NK receptor antagonist) and GR82334-(an NK1 antagonist) sensitive increase in inspiratory burst amplitude recorded from XII nerves. 3. Under current clamp, SP(NK1) significantly depolarized XII MNs, potentiated repetitive firing responses to injected currents and produced a leftward shift in the firing frequency-current relationships without affecting slope. 4. Under voltage clamp, SP(NK1) evoked an inward current and increased input resistance, but had no effect on inspiratory synaptic currents. SP(NK1) currents persisted in the presence of TTX, were GR82334 sensitive, were reduced with hyperpolarization and reversed near the expected E(K). 5. Effects of the alpha(1)-noradrenergic receptor agonist phenylephrine (PE) on repetitive firing behaviour were virtually identical to those of SP(NK1). Moreover, SP(NK1) currents were completely occluded by PE, suggesting that common intracellular pathways mediate the actions of NK1 and alpha(1)-noradrenergic receptors. In spite of the similar actions of SP(NK1) and PE on XII MN responses to somally injected current, alpha(1)-noradrenergic receptor activation potentiated inspiratory synaptic currents and was more than twice as effective in potentiating XII nerve inspiratory burst amplitude. 6. GR82334 reduced XII nerve inspiratory burst amplitude and generated a small outward current in XII MNs. These observations, together with the first immunohistochemical evidence in the newborn for SP immunopositive terminals in the vicinity of SP(NK1)-sensitive inspiratory XII MNs, support the endogenous modulation of XII MN excitability by SP. 7. In contrast to phrenic MNs (Ptak et al. 2000), blocking NMDA receptors with AP5 had no effect on the modulation of XII nerve activity by SP(NK1). 8. In conclusion, SP(NK1) modulates XII motoneuron responses to inspiratory drive primarily through inhibition of a resting, postsynaptic K+ leak conductance. The results establish the functional significance of SP in controlling upper airway tone during early postnatal life and indicate differential modulation of motoneurons controlling airway and pump muscles by SP.


Subject(s)
Hypoglossal Nerve/cytology , Hypoglossal Nerve/physiology , Motor Neurons/physiology , Physalaemin/analogs & derivatives , Receptors, Neurokinin-1/physiology , Animals , Animals, Newborn , Hypoglossal Nerve/chemistry , Immunohistochemistry , In Vitro Techniques , Mice , Mice, Inbred Strains , Patch-Clamp Techniques , Phenylephrine/pharmacology , Physalaemin/pharmacology , Potassium/pharmacology , Respiratory Center/physiology , Respiratory Mechanics/physiology , Substance P/analysis , Substance P/pharmacology , Sympathomimetics/pharmacology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Tetrodotoxin/pharmacology
20.
J Urol ; 165(5): 1739-44, 2001 May.
Article in English | MEDLINE | ID: mdl-11342967

ABSTRACT

PURPOSE: Spinal cord injury above the sacral micturition center usually leads to detrusor hyperreflexia, increased intravesical pressure and post-void residual urine. Detrusor hyperreflexia is believed to be mediated by afferent C fibers with tachykinins as neurotransmitters. We investigated the selective peptide tachykinin antagonists MEN 11420 and GR 82334 of NK-2 and NK-1 receptors, respectively, in a chronic rat model of detrusor hyperreflexia after suprasacral spinal cord injury. MATERIALS AND METHODS: Adult female Sprague-Dawley rats weighing 200 to 250 gm. were used. The spinal cord was transected at the T10 level. The bladder was evacuated by the Credé maneuver 3 times daily. After 6 weeks the rats were implanted with femoral vein and bladder dome catheters 2 days before filling cystometry. The 5 rats in group 1 received 100 nmol./kg. of the NK-2 antagonist MEN 11420 intravenously. The 5 rats in group 2 received 100 nmol./kg. of the NK-1 antagonist GR 82334 intravenously. The 5 rats in group 3 received a combination of the same dose of each antagonist. Three repetitive micturition cycles were recorded before injection. Three micturition cycles were done 20 minutes after the injection of each antagonist. Mean cystometric parameters were reported, including bladder capacity, micturition pressure, baseline pressure, post-void residual urine and micturition volume, and the number and amplitude of hyperreflexic contractions greater than 15 cm. water. RESULTS: MEN 11420 significantly reduced the frequency of hyperreflexic contractions and baseline bladder pressure (p <0.05). There was no statistically significant effect on the other cystometric parameters. GR 82334 reduced the amplitude of hyperreflexic contractions but not statistically significant. A combination of MEN 11420 and GR 82334 significantly reduced the frequency and amplitude of hyperreflexic contractions (p <0.05) with no significant effects on other cystometric parameters, although there was a tendency toward increased micturition volume and bladder capacity. CONCLUSIONS: These results suggest that at the peripheral level there is an efferent role of tachykinins in detrusor hyperreflexia after spinal cord injury. NK-1 and NK-2 receptor selective antagonists reduced the frequency and amplitude of hyperreflexic contractions as well as baseline bladder pressure. This finding may lead to potential new therapeutic modalities using selective tachykinins antagonists with other pharmacological agents to combat detrusor hyperreflexia.


Subject(s)
Neurokinin-1 Receptor Antagonists , Peptides, Cyclic/pharmacology , Receptors, Neurokinin-2/antagonists & inhibitors , Spinal Cord Injuries/complications , Tachykinins/physiology , Urinary Bladder, Neurogenic/physiopathology , Urinary Bladder/physiopathology , Animals , Female , Muscle Contraction/drug effects , Physalaemin/analogs & derivatives , Physalaemin/pharmacology , Rats , Rats, Sprague-Dawley , Reflex, Abnormal/drug effects , Spinal Cord Injuries/physiopathology , Urinary Bladder/drug effects , Urinary Bladder, Neurogenic/etiology , Urodynamics
SELECTION OF CITATIONS
SEARCH DETAIL
...