Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 635
Filter
1.
Mol Neurobiol ; 58(11): 5772-5789, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34406600

ABSTRACT

Increasing evidence suggests that transmembrane protein 16A (TMEM16A) in nociceptive neurons is an important molecular component contributing to peripheral pain transduction. The present study aimed to evaluate the role and mechanism of TMEM16A in chronic nociceptive responses elicited by spared nerve injury (SNI). In this study, SNI was used to induce neuropathic pain. Drugs were administered intrathecally. The expression and cellular localization of TMEM16A, the ERK pathway, and NK-1 in the dorsal root ganglion (DRG) were detected by western blot and immunofluorescence. Behavioral tests were used to evaluate the role of TMEM16A and p-ERK in SNI-induced persistent pain and hypersensitivity. The role of TMEM16A in the hyperexcitability of primary nociceptor neurons was assessed by electrophysiological recording. The results show that TMEM16A, p-ERK, and NK-1 are predominantly expressed in small neurons associated with nociceptive sensation. TMEM16A is colocalized with p-ERK/NK-1 in DRG. TMEM16A, the MEK/ERK pathway, and NK-1 are activated in DRG after SNI. ERK inhibitor or TMEM16A antagonist prevents SNI-induced allodynia. ERK and NK-1 are downstream of TMEM16A activation. Electrophysiological recording showed that CaCC current increases and intrathecal application of T16Ainh-A01, a selective TMEM16A inhibitor, reverses the hyperexcitability of DRG neurons harvested from rats after SNI. We conclude that TMEM16A activation in DRG leads to a positive interaction of the ERK pathway with activation of NK-1 production and is involved in the development of neuropathic pain after SNI. Also, the blockade of TMEM16A or inhibition of the downstream ERK pathway or NK-1 upregulation may prevent the development of neuropathic pain.


Subject(s)
Anoctamins/physiology , Extracellular Signal-Regulated MAP Kinases/physiology , Ganglia, Spinal/pathology , Hyperalgesia/physiopathology , Neuralgia/physiopathology , Peroneal Nerve/injuries , Receptors, Neurokinin-1/physiology , Sensory Receptor Cells/physiology , Signal Transduction/physiology , Tibial Nerve/injuries , Animals , Anoctamins/antagonists & inhibitors , Butadienes/pharmacology , Chronic Pain/etiology , Chronic Pain/physiopathology , Hyperalgesia/etiology , Ligation , Male , Neuralgia/etiology , Nitriles/pharmacology , Nociception/physiology , Pyrimidines/pharmacology , Random Allocation , Rats , Rats, Sprague-Dawley , Thiazoles/pharmacology
2.
Neuropharmacology ; 196: 108716, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34273385

ABSTRACT

The basal ganglia are a group of sub-cortical structures believed to play a critical role in action selection and sequencing. The striatum is the largest input structure of the basal ganglia and contains the neuropeptide substance P in abundance. Recent computational work has suggested that substance P could play a critical role in action sequence performance and acquisition, but this has not been tested experimentally before. The aim of the present study was to test how blocking substance P's main NK1-type receptors affected the sequential and temporal organization of spontaneous behavioral patterns. We did this in rats by focusing on the grooming chain, an innate and highly stereotyped ordered sequence. We performed an open field experiment in which the NK1 receptor antagonist L-733,060 was injected intraperitoneally in rats at two doses (2 and 4 mg/kg/ml), in a within-subject counterbalanced design. We used first order transition probabilities, Variable Length Markov Models, entropy metrics and T-pattern analysis to evaluate the effects of L-733,060 on sequential and temporal aspects of spontaneously ordered behavioral sequences. Our results suggest that blocking NK1 receptors made the transitions between the grooming chain elements significantly more variable, the transition structure of the grooming bouts simpler, and it increased the probability of transitioning from active to inactive states. Overall, this suggest that blocking substance P receptors led to a general break down in the fluency of spontaneous behavioral sequences, suggesting that substance P could be playing a key role in the implementation of sequential patterns.


Subject(s)
Behavior, Animal/drug effects , Grooming/drug effects , Neurokinin-1 Receptor Antagonists/pharmacology , Piperidines/pharmacology , Receptors, Neurokinin-1/physiology , Substance P/physiology , Animals , Basal Ganglia , Grooming/physiology , Markov Chains , Rats
4.
Addict Biol ; 25(6): e12822, 2020 11.
Article in English | MEDLINE | ID: mdl-31830773

ABSTRACT

The opioid epidemic has become a severe public health problem, with approximately 130 opioid-induced deaths occurring each day in the United States. Prescription opioids are responsible for approximately 40% of these deaths. Oxycodone is one of the most commonly abused prescription opioids, but despite its prevalent misuse, the number of preclinical studies investigating oxycodone-seeking behaviors is relatively limited. Furthermore, preclinical oxycodone studies that include female subjects are even more scarce, and it is critical that future work includes both sexes. Additionally, the oral route of administration is one of the most common routes for recreational users, especially in the early stages of drug experimentation. However, currently, only two studies have been published investigating operant oral oxycodone self-administration in rodents. Therefore, the primary goal of the present study was to establish an oral oxycodone operant self-administration model in adult male and female rats, as well as to examine a potential mechanism of stress-primed reinstatement. We found that females consumed significantly more oral oxycodone than males in operant self-administration sessions. We also found that active oxycodone self-administration was reduced by mu opioid receptor antagonism and by substitution of water for oxycodone solution. Lastly, we induced stress-primed reinstatement and found that this behavior was significantly attenuated by antagonism of the neurokinin-1 receptor, consistent with our prior work examining stress-induced reinstatement of alcohol- and cocaine-seeking.


Subject(s)
Analgesics, Opioid/administration & dosage , Oxycodone/administration & dosage , Prescription Drug Misuse , Receptors, Neurokinin-1/physiology , Self Administration , Analgesics, Opioid/blood , Animals , Drug-Seeking Behavior/drug effects , Estrous Cycle/drug effects , Extinction, Psychological/drug effects , Female , Male , Naloxone/administration & dosage , Neurokinin-1 Receptor Antagonists/pharmacology , Oxycodone/blood , Rats , Rats, Wistar , Receptors, Opioid, mu/antagonists & inhibitors
6.
Eur J Pharmacol ; 865: 172806, 2019 Dec 15.
Article in English | MEDLINE | ID: mdl-31738934

ABSTRACT

Δ9-THC suppresses cisplatin-induced vomiting through activation of cannabinoid CB1 receptors. Cisplatin-evoked emesis is predominantly due to release of serotonin and substance P (SP) in the gut and the brainstem which subsequently stimulate their corresponding 5-HT3-and neurokinin NK1-receptors to induce vomiting. Δ9-THC can inhibit vomiting caused either by the serotonin precursor 5-HTP, or the 5-HT3 receptor selective agonist, 2-methyserotonin. In the current study, we explored whether Δ9-THC and related CB1/CB2 receptor agonists (WIN55,212-2 and CP55,940) inhibit vomiting evoked by SP (50 mg/kg, i.p.) or the NK1 receptor selective agonist GR73632 (5 mg/kg, i.p.). Behavioral methods were employed to determine the antiemetic efficacy of cannabinoids in least shrews. Our results showed that administration of varying doses of Δ9-THC (i.p. or s.c.), WIN55,212-2 (i.p.), or CP55,940 (i.p.) caused significant suppression of SP-evoked vomiting in a dose-dependent manner. When tested against GR73632, Δ9-THC also dose-dependently reduced the evoked emesis. The antiemetic effect of Δ9-THC against SP-induced vomiting was prevented by low non-emetic doses of the CB1 receptor inverse-agonist/antagonist SR141716A (<10 mg/kg). We also found that the NK1 receptor antagonist netupitant can significantly suppress vomiting caused by a large emetic dose of SR141716A (20 mg/kg). In sum, Δ9-THC and related cannabinoids suppress vomiting evoked by the nonselective (SP) and selective (GR73632) neurokinin NK1 receptor agonists via stimulation of cannabinoid CB1 receptors.


Subject(s)
Benzoxazines/therapeutic use , Cannabinoid Receptor Agonists/therapeutic use , Cannabinoids/therapeutic use , Cyclohexanols/therapeutic use , Dronabinol/therapeutic use , Morpholines/therapeutic use , Naphthalenes/therapeutic use , Receptors, Neurokinin-1/physiology , Vomiting/drug therapy , Animals , Female , Male , Peptide Fragments/pharmacology , Shrews , Substance P/analogs & derivatives , Substance P/pharmacology , Vomiting/chemically induced
7.
J Physiol Biochem ; 75(4): 415-421, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31372898

ABSTRACT

Tachykinins (TKs) include an evolutionarily conserved group of small bio-active peptides which possess a common carboxyl-terminal sequence, Phe-X-Gly-Leu-Met-NH2. TKs also have been shown to have implications in different steps of carcinogenesis, such as angiogenesis, mitogenesis, metastasis, and other growth-related events. The biological actions of substance P (SP), as the most important member of the TK family, are mainly mediated through a G protein-coupled receptor named neurokinin-1 receptor (NK1R). More recently, it has become clear that SP/NK1R system is involved in the initiation and activation of signaling pathways involved in cancer development and progression. Therefore, SP may contribute to triggering a variety of effector mechanisms including protein synthesis and a number of transcription factors that modulate the expression of genes involved in these processes. The overwhelming insights into the blockage of NK1R using specific antagonists could suggest a therapeutic approach in cancer therapy. In this review, we focus on evidence supporting an association between the signaling pathways of the SP/NK1R system and cancer cell proliferation and development.


Subject(s)
Carcinogenesis/metabolism , Neoplasms/metabolism , Receptors, Neurokinin-1/physiology , Substance P/physiology , Animals , Cell Line, Tumor , Cell Proliferation , Humans , Signal Transduction
8.
Neuron ; 103(3): 432-444.e3, 2019 08 07.
Article in English | MEDLINE | ID: mdl-31221559

ABSTRACT

Subtypes of nucleus accumbens medium spiny neurons (MSNs) promote dichotomous outcomes in motivated behaviors. However, recent reports indicate enhancing activity of either nucleus accumbens (NAc) core MSN subtype augments reward, suggesting coincident MSN activity may underlie this outcome. Here, we report a collateral excitation mechanism in which high-frequency, NAc core dopamine 1 (D1)-MSN activation causes long-lasting potentiation of excitatory transmission (LLP) on dopamine receptor 2 (D2)-MSNs. Our mechanistic investigation demonstrates that this form of plasticity requires release of the excitatory peptide substance P from D1-MSNs and robust cholinergic interneuron activation through neurokinin receptor stimulation. We also reveal that D2-MSN LLP requires muscarinic 1 receptor activation, intracellular calcium signaling, and GluR2-lacking AMPAR insertion. This study uncovers a mechanism for shaping NAc core activity through the transfer of excitatory information from D1-MSNs to D2-MSNs and may provide a means for altering goal-directed behavior through coordinated MSN activity.


Subject(s)
Dopaminergic Neurons/physiology , Long-Term Potentiation/physiology , Nucleus Accumbens/physiology , Substance P/metabolism , Action Potentials/physiology , Animals , Aprepitant/pharmacology , Calcium Signaling/physiology , Cholinergic Neurons/physiology , Dopaminergic Neurons/radiation effects , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Interneurons/physiology , Mice , Mice, Inbred C57BL , Motivation , Neurokinin-1 Receptor Antagonists/pharmacology , Nucleus Accumbens/cytology , Photic Stimulation , Piperidines/pharmacology , Receptor, Muscarinic M1/physiology , Receptors, AMPA/physiology , Receptors, Dopamine D1/analysis , Receptors, Dopamine D2/analysis , Receptors, Neurokinin-1/physiology
9.
eNeuro ; 6(3)2019.
Article in English | MEDLINE | ID: mdl-31122949

ABSTRACT

Reelin (Reln) and Disabled-1 (Dab1) participate in the Reln-signaling pathway and when either is deleted, mutant mice have the same spinally mediated behavioral abnormalities, increased sensitivity to noxious heat and a profound loss in mechanical sensitivity. Both Reln and Dab1 are highly expressed in dorsal horn areas that receive and convey nociceptive information, Laminae I-II, lateral Lamina V, and the lateral spinal nucleus (LSN). Lamina I contains both projection neurons and interneurons that express Neurokinin-1 receptors (NK1Rs) and they transmit information about noxious heat both within the dorsal horn and to the brain. Here, we ask whether the increased heat nociception in Reln and dab1 mutants is due to incorrectly positioned dorsal horn neurons that express NK1Rs. We found more NK1R-expressing neurons in Reln-/- and dab1-/- Laminae I-II than in their respective wild-type mice, and some NK1R neurons co-expressed Dab1 and the transcription factor Lmx1b, confirming their excitatory phenotype. Importantly, heat stimulation in dab1-/- mice induced Fos in incorrectly positioned NK1R neurons in Laminae I-II. Next, we asked whether these ectopically placed and noxious-heat responsive NK1R neurons participated in pain behavior. Ablation of the superficial NK1Rs with an intrathecal injection of a substance P analog conjugated to the toxin saporin (SSP-SAP) eliminated the thermal hypersensitivity of dab1-/- mice, without altering their mechanical insensitivity. These results suggest that ectopically positioned NK1R-expressing neurons underlie the heat hyperalgesia of Reelin-signaling pathway mutants, but do not contribute to their profound mechanical insensitivity.


Subject(s)
Cell Adhesion Molecules, Neuronal/physiology , Extracellular Matrix Proteins/physiology , Hyperalgesia/physiopathology , Nerve Tissue Proteins/physiology , Posterior Horn Cells/physiology , Receptors, Neurokinin-1/physiology , Serine Endopeptidases/physiology , Animals , Cell Adhesion Molecules, Neuronal/genetics , Extracellular Matrix Proteins/genetics , Hot Temperature , Male , Mice, Knockout , Nerve Tissue Proteins/genetics , Posterior Horn Cells/metabolism , Receptors, Neurokinin-1/metabolism , Reelin Protein , Serine Endopeptidases/genetics , Signal Transduction , Spinal Cord/physiopathology
10.
Cereb Cortex ; 29(3): 1090-1108, 2019 03 01.
Article in English | MEDLINE | ID: mdl-29462275

ABSTRACT

We have proposed that cortical nNOS/NK1R interneurons have a role in sleep homeostasis. The hypocretins (orexins) are wake-promoting neuropeptides and hypocretin/orexin (Hcrt) neurons project to the cortex. Hcrt peptides affect deep layer cortical neurons, and Hcrt receptor 1 (Hcrtr1; Ox1r) mRNA is expressed in cortical nNOS/NK1R cells. Therefore, we investigated whether Hcrt neuron stimulation affects cingulate cortex nNOS/NK1R neurons. Bath application of HCRT1/orexin-A evoked an inward current and membrane depolarization in most nNOS/NK1R cells which persisted in tetrodotoxin; optogenetic stimulation of Hcrt terminals expressing channelrhodopsin-2 confirmed these results, and pharmacological studies determined that HCRTR1 mediated these responses. Single-cell RT-PCR found Hcrtr1 mRNA in 31% of nNOS/NK1R cells without any Hcrtr2 mRNA expression; immunohistochemical studies of Hcrtr1-EGFP mice confirmed that a minority of nNOS/NK1R cells express HCRTR1. When Hcrt neurons degenerated in orexin-tTA;TetO DTA mice, the increased EEG delta power during NREM sleep produced in response to 4 h sleep deprivation and c-FOS expression in cortical nNOS/NK1R cells during recovery sleep were indistinguishable from that of controls. We conclude that Hcrt excitatory input to these deep layer cells is mediated through HCRTR1 but is unlikely to be involved in the putative role of cortical nNOS/NK1R neurons in sleep homeostasis.


Subject(s)
Gyrus Cinguli/physiology , Homeostasis , Neurons/physiology , Nitric Oxide Synthase Type I/physiology , Orexin Receptors/physiology , Receptors, Neurokinin-1/physiology , Sleep/physiology , Animals , Female , Gyrus Cinguli/drug effects , Hypothalamic Area, Lateral/physiology , Male , Mice, Inbred C57BL , Neurons/drug effects , Orexins/administration & dosage , Orexins/physiology
11.
Exp Eye Res ; 177: 87-95, 2018 12.
Article in English | MEDLINE | ID: mdl-30076797

ABSTRACT

PURPOSE: to explore whether the NK1 and Y2 receptors are involved in the pathogenesis of laser-induced CNV (choroidal neovascularization) in C57Bl/6N mice. METHODS: CNV was induced by laser damage of Bruch's membrane and the CNV volume was determined by OCT and/or flatmount preparation. First, the development of the CNV volume over time was evaluated. Second, the CNV development in NK1- and Y2 KO mice was analyzed. Third, the effect on the development as well as the regression of CNV by intravitreal injections of the NK1 antagonist SR140333 and the Y2 antagonist BIIEO246 separately and each in combination with Eylea®, was investigated. Furthermore, flatmount CNV volume measurements were correlated to volumes obtained by the in vivo OCT technique. RESULTS: CNV volume peak was observed at day 4 after laser treatment. Compared to wild type mice, NK1 and Y2 KO mice showed significantly smaller CNV volumes. Eylea® and the Y2 antagonist significantly reduced the volume of the developing CNV. In contrast to Eylea® there was no effect of either antagonist on the regression of CNV, additionally no additive effect upon combined Eylea®/antagonist treatment was observed. There was a strong positive correlation between CNV volumes obtained by OCT and flatmount. CONCLUSION: NK1 and Y2 receptors mediate the development of laser-induced CNVs in mice. They seem to play an important role at the developmental stage of CNVs, whereas VEGF via VEGF receptor may be an important mediator throughout the CNV existence. In vivo OCT correlates with flatmount CNV volume, representing a useful tool for in vivo evaluations of CNV over time.


Subject(s)
Choroidal Neovascularization , Receptors, Neurokinin-1/physiology , Receptors, Neuropeptide Y/physiology , Angiogenesis Inhibitors/pharmacology , Animals , Cells, Cultured , Choroid/pathology , Choroidal Neovascularization/drug therapy , Choroidal Neovascularization/pathology , Choroidal Neovascularization/physiopathology , Disease Models, Animal , Endothelial Cells/drug effects , Fluorescein Angiography , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurokinin-1 Receptor Antagonists/pharmacology , Receptors, Neurokinin-1/deficiency , Receptors, Neuropeptide Y/antagonists & inhibitors , Receptors, Neuropeptide Y/deficiency , Receptors, Vascular Endothelial Growth Factor , Recombinant Fusion Proteins/pharmacology
12.
Br J Pharmacol ; 175(15): 3162-3174, 2018 08.
Article in English | MEDLINE | ID: mdl-29767468

ABSTRACT

BACKGROUND AND PURPOSE: The adenosine A1 receptor is reported to mediate several excitatory effects in the airways and has inhibitory effects in the CNS. In this study, we investigated the role of peripheral and central A1 receptors in regulating cough and airway obstruction. EXPERIMENTAL APPROACH: Drugs were administered to guinea pigs via inhalation or i.c.v. infusion. Following the administration of different drugs, cough was induced by exposing guinea pigs to aerosolized 0.4 M citric acid. An automated analyser recorded both cough and airway obstruction simultaneously using whole-body plethysmography. KEY RESULTS: The A1 receptor agonist, cyclopentyladenosine (CPA, administered by inhalation), dose-dependently inhibited cough and also inhibited airway obstruction. Similarly, CPA, administered i.c.v., inhibited both the citric acid-induced cough and airway obstruction; this was prevented by pretreatment with the A1 receptor antagonist DPCPX (i.c.v.). Treatment with DPCPX alone dose-dependently enhanced the citric acid-induced cough and airway obstruction. This effect was reversed following treatment with either the glutamate GluN1 receptor antagonist D-AP5 or the neurokinin NK1 receptor antagonist FK-888. CONCLUSIONS AND IMPLICATIONS: These findings suggest that activation of either peripheral or central adenosine A1 receptors inhibits citric acid-induced cough and airway obstruction. The data also suggest that tonic activation of central adenosine A1 receptors serves as a negative regulator of cough and airway obstruction, secondary to inhibition of excitatory glutamatergic and tachykininergic neurotransmission.


Subject(s)
Airway Obstruction/physiopathology , Cough/physiopathology , Receptor, Adenosine A1/physiology , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine A1 Receptor Agonists/pharmacology , Adenosine A1 Receptor Antagonists/pharmacology , Administration, Inhalation , Animals , Citric Acid/administration & dosage , Female , Guinea Pigs , Male , Receptors, Neurokinin-1/physiology , Synaptic Transmission , Xanthines/pharmacology , gamma-Aminobutyric Acid/pharmacology
13.
Neurobiol Learn Mem ; 148: 60-68, 2018 02.
Article in English | MEDLINE | ID: mdl-29519453

ABSTRACT

A single threatening experience may change the behavior of an animal in a long-lasting way and elicit generalized behavioral responses to a novel threatening situation that is unrelated to the original aversive experience. Electrical stimulation (ES) of the dorsal periaqueductal gray (dPAG) produces a range of defensive reactions, characterized by freezing, escape, and post-stimulation freezing (PSF). The latter reflects the processing of ascending aversive information to prosencephalic structures, including the central nucleus of the amygdala (CeA), which allows the animal to evaluate the consequences of the aversive situation. This process is modulated by substance P (SP) and its preferred receptor, neurokinin 1 (NK1). The ventral hippocampus (VH) has been associated with the processing of aversive information and expression of emotional reactions with negative valence, but the participation of the VH in the expression of these defensive responses has not been investigated. The VH is rich in NK1 receptor expression and has a high density of SP-containing fibers. The present study examined the role of NK1 receptors in the VH in the expression of defensive responses and behavioral sensitization that were induced by dPAG-ES. Rats were implanted with an electrode in the dPAG for ES, and a cannula was implanted in the VH or CeA for injections of vehicle (phosphate-buffered saline) or the NK1 receptor antagonist spantide (100 pmol/0.2 µL. Spantide reduced the duration of PSF that was evoked by dPAG-ES, without changing the aversive freezing or escape thresholds. One and 7 days later, exploratory behavior was evaluated in independent groups of rats in the elevated plus maze (EPM). dPAG-ES in rats that received vehicle caused higher aversion to the open arms of the EPM compared with rats that did not receive dPAG stimulation at both time intervals. Injections of spantide in the VH or CeA prevented the proaversive effects of dPAG-ES in the EPM only 1 day later. These findings suggest that NK1 receptors are activated in both the VH and CeA during the processing of aversive information that derives from dPAG-ES. As previously shown for the CeA, SP/NK1 receptors in the VH are recruited during PSF that is evoked by dPAG-ES, suggesting that a 24-h time window is susceptible to interventions with NK1 antagonists that block the passage of aversive information from the dPAG to higher brain areas.


Subject(s)
Avoidance Learning , Behavior, Animal , Central Amygdaloid Nucleus/drug effects , Central Nervous System Sensitization , Exploratory Behavior , Hippocampus/drug effects , Maze Learning , Neurokinin-1 Receptor Antagonists/pharmacology , Periaqueductal Gray , Receptors, Neurokinin-1/physiology , Substance P/analogs & derivatives , Animals , Avoidance Learning/drug effects , Avoidance Learning/physiology , Behavior, Animal/drug effects , Behavior, Animal/physiology , Central Nervous System Sensitization/drug effects , Central Nervous System Sensitization/physiology , Electric Stimulation , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Male , Maze Learning/drug effects , Maze Learning/physiology , Neurokinin-1 Receptor Antagonists/administration & dosage , Periaqueductal Gray/physiology , Rats , Rats, Wistar , Substance P/administration & dosage , Substance P/pharmacology
14.
Naunyn Schmiedebergs Arch Pharmacol ; 391(3): 299-308, 2018 03.
Article in English | MEDLINE | ID: mdl-29279967

ABSTRACT

Tachykinin NK2 receptor (NK2R) agonists have potential to alleviate clinical conditions associated with bladder and gastrointestinal under activity. The effects of agonists with differing selectivity for NK2R over NK1Rs on colorectal, bladder, and cardiovascular function were examined in anesthetized dogs. Intravenous (IV) administration of NKA, LMN-NKA ([Lys5,MeLeu9,Nle10]-NKA(4-10)), and [ß-Ala8]-NKA(4-10) caused a dose-related increase in colorectal pressure (up to 98 mmHg) that was blocked by pretreatment with the NK2R antagonist GR 159897 (1 mg/kg), and hypotension (decrease in mean arterial pressure of ~40 mmHg) that was blocked by the NK1R antagonist CP-99,994 (1 mg/kg). Despite the greater in vitro selectivity of LMN-NKA and [ß-Ala8]-NKA(4-10) for NK2R over NK1Rs compared with NKA, all 3 agonists increased colorectal pressure and caused hypotension within a similar dose range when administered as a bolus (0.1-300 µg/kg IV), or even as a slow IV infusion over 5 min (NKA; 0.02-0.6 µg/kg/min). In contrast, subcutaneous (SC) administration of LMN-NKA (3-10 µg/kg) increased colorectal pressure (up to 50 mmHg) and elicited micturition (≧ 85% voiding efficiency) without causing hypotension. NK2R agonists can produce rapid-onset, short-duration, colorectal contractions, and efficient voiding of urine without hypotension after SC administration, indicating that routes of administration that avoid the high plasma concentrations associated with IV dosing improve the separation between desired and unwanted pharmacodynamic effects. The potent hypotensive effect of NKA in dogs was unexpected based on published studies in humans in which IV infusion of NKA did not affect blood pressure at doses that increased gastrointestinal motility.


Subject(s)
Arterial Pressure/drug effects , Colon/drug effects , Neurokinin A/analogs & derivatives , Neurokinin A/pharmacology , Receptors, Neurokinin-1/physiology , Receptors, Neurokinin-2/physiology , Urinary Bladder/drug effects , Anesthesia , Animals , Colon/physiology , Dogs , Female , Heart Rate/drug effects , Indoles/pharmacology , Male , Neurokinin-1 Receptor Antagonists/pharmacology , Piperidines/pharmacology , Receptors, Neurokinin-2/agonists , Receptors, Neurokinin-2/antagonists & inhibitors , Urinary Bladder/physiology
15.
Int Rev Neurobiol ; 136: 151-175, 2017.
Article in English | MEDLINE | ID: mdl-29056150

ABSTRACT

Substance P (SP) is an 11-amino acid neuropeptide of the tachykinin family that preferentially activates the neurokinin-1 receptor (NK1R). First isolated 85 years ago and sequenced 40 years later, SP has been extensively studied. Early studies identified a role for SP and the NK1R in contraction of intestinal smooth muscle, central pain processing, and neurogenic inflammation. An FDA-approved NK1R antagonist, aprepitant, is used clinically for the treatment of chemotherapy-induced nausea, as the NK1R influences the activity of the brain stem emesis centers. More recently, SP and the NK1R have gained attention for their role in complex psychiatric processes including stress, anxiety, and depression. However, clinical development of NK1R antagonists for these indications has so far been unsuccessful. Several preclinical studies have also demonstrated a role of the NK1R in drug taking and drug seeking, especially as it relates to escalated consumption and stress-elicited seeking. This line of research developed in parallel with findings supporting a role of corticotropin-releasing factor (CRF) in stress-induced drug seeking. Over this time, CRF arguably gained more attention as a target for development of addiction pharmacotherapies. However, this effort has not resulted in a viable drug for use in human populations. Given promising clinical findings for the efficacy of NK1R antagonists on craving in alcoholics, along with recent data suggesting that a number of negative results from NK1R trials were likely due to insufficient receptor occupancy, the NK1R merits being revisited as a target for the development of novel pharmacotherapeutics for addiction.


Subject(s)
Mood Disorders/metabolism , Neurokinin-1 Receptor Antagonists/therapeutic use , Receptors, Neurokinin-1/physiology , Substance P/physiology , Substance-Related Disorders/metabolism , Animals , Humans , Mood Disorders/drug therapy , Receptors, Neurokinin-1/metabolism , Substance P/metabolism , Substance-Related Disorders/drug therapy
16.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 33(8): 1108-1112, 2017 Aug.
Article in Chinese | MEDLINE | ID: mdl-28871952

ABSTRACT

Objective To investigate the expressions of substance P (SP) and neurokinin-1 receptor (NK1R) in eosinophil-enriched blood cells from patients with chronic spontaneous urticaria (CSU). Methods Peripheral venous blood samples were collected from patients with CSU and healthy controls (HCs), and then stimulated with crude extracts of Artemisia pollen, dust mite, and Platanus pollen (all at concentrations of 0.1 and 1.0 µg/mL). The expressions of SP and NK1R in eosinophil-enriched blood cells were detected by flow cytometry. Results Compared with HCs, eosinophil proportion in peripheral blood of CSU patients increased 1.2-fold. Percentage of NK1R+ eosinophils in the patients with CSU was elevated up to 66% compared with HCs when cultured in the medium only. However, the level of SP decreased by 40% in the CSU patients. In eosinophil-enriched blood cells from the CSU patients, the crude extract of dust mite at 0.1 µg/mL induced approximately 1.11-fold increase of NK1R expression. Conclusion Expression of NK1R increases in the eosinophils of CSU patients. Blockers of NK1R might be used for CSU treatment.


Subject(s)
Eosinophilia/etiology , Receptors, Neurokinin-1/physiology , Urticaria/etiology , Adult , Animals , Chronic Disease , Female , Humans , Male , Middle Aged , Neurokinin-1 Receptor Antagonists/therapeutic use , Pollen/immunology , Pyroglyphidae/immunology , Receptors, Neurokinin-1/blood , Substance P/blood , Up-Regulation , Urticaria/blood , Urticaria/drug therapy
17.
Curr Opin Pharmacol ; 35: 94-100, 2017 08.
Article in English | MEDLINE | ID: mdl-28803835

ABSTRACT

Receptor distribution studies have played a key role in the characterization of receptor systems (e.g. GABAB, NMDA (GluNRs), and Neurokinin 1) and in generating hypotheses to exploit these systems as potential therapeutic targets. Distribution studies can provide important information on the potential role of candidate receptors in normal physiology/disease and alert for possible adverse effects of targeting the receptors. Moreover, they can provide valuable information relating to quantitative target engagement (e.g. % receptor occupancy) to drive mechanistic pharmacokinetic/pharmacodynamic (PK/PD) hypotheses for compounds in the Drug Discovery process. Finally, receptor distribution and quantitative target engagement studies can be used to validate truly translational technologies such as PET ligands and pharmacoEEG paradigms to facilitate bridging of the preclinical/clinical interface and thus increase probability of success.


Subject(s)
Receptors, GABA-B/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Receptors, Neurokinin-1/physiology , Animals , Humans
18.
Eur J Pain ; 21(7): 1277-1284, 2017 08.
Article in English | MEDLINE | ID: mdl-28493529

ABSTRACT

BACKGROUND: Substance P (SP) is a pain- and inflammation-related neuropeptide which preferentially binds to the neurokinin receptor 1 (NK1 ). SP and NK1 receptors have been implicated in joint pain, inflammation and damage in animal models and human studies of osteoarthritis (OA). The aim of this study was to test if genetic variation at the neurokinin 1 receptor gene (TACR1) is associated with pain in individuals with radiographic knee OA. METHODS: Participants from the Genetics of OA and Lifestyle study were used for the discovery group (n = 1615). Genotype data for six SNPs selected to cover most variation in the TACR1 gene were used to test for an association with symptomatic OA. Replication analysis was performed using data from the Chingford 1000 Women Study, Hertfordshire Cohort Study, Tasmanian Older Adult Cohort Study and the Clearwater OA Study. In total, n = 1715 symptomatic OA and n = 735 asymptomatic OA individuals were analysed. RESULTS: Out of six SNPs tested in the TACR1 gene, one (rs11688000) showed a nominally significant association with a decreased risk of symptomatic OA in the discovery cohort. This was then replicated in four additional cohorts. After adjusting for age, gender, body mass index and radiographic severity, the G (minor) allele at rs11688000 was associated with a decreased risk of symptomatic OA compared to asymptomatic OA cases (p = 9.90 × 10-4 , OR = 0.79 95% 0.68-0.90 after meta-analysis). CONCLUSIONS: This study supports a contribution from the TACR1 gene in human OA pain, supporting further investigation of this gene's function in OA. SIGNIFICANCE: This study contributes to the knowledge of the genetics of painful osteoarthritis, a condition which affects millions of individuals worldwide. Specifically, a contribution from the TACR1 gene to modulating pain sensitivity in osteoarthritis is suggested.


Subject(s)
Arthralgia/physiopathology , Genetic Variation/genetics , Osteoarthritis, Knee/physiopathology , Pain/genetics , Polymorphism, Single Nucleotide/physiology , Receptors, Neurokinin-1/chemistry , Substance P/chemistry , Animals , Cohort Studies , Female , Genotype , Humans , Pain/physiopathology , Phenotype , Receptors, Neurokinin-1/physiology
19.
J Immunol ; 197(10): 4021-4033, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27798158

ABSTRACT

Substance P neuropeptide and its receptor, neurokinin-1 receptor (NK1R), are reported to present on the ocular surface. In this study, mice lacking functional NK1R exhibited an excessive desquamation of apical corneal epithelial cells in association with an increased epithelial cell proliferation and increased epithelial cell density, but decreased epithelial cell size. The lack of NK1R also resulted in decreased density of corneal nerves, corneal epithelial dendritic cells (DCs), and a reduced volume of basal tears. Interestingly, massive accumulation of CD11c+CD11b+ conventional DCs was noted in the bulbar conjunctiva and near the limbal area of corneas from NK1R-/- mice. After ocular HSV-1 infection, the number of conventional DCs and neutrophils infiltrating the infected corneas was significantly higher in NK1R-/- than C57BL/6J mice. This was associated with an increased viral load in infected corneas of NK1R-/- mice. As a result, the number of IFN-γ-secreting virus-specific CD4 T cells in the draining lymph nodes of NK1R-/- mice was much higher than in infected C57BL/6J mice. An increased number of CD4 T cells and mature neutrophils (CD11b+Ly6ghigh) in the inflamed corneas of NK1R-/- mice was associated with an early development of severe herpes stromal keratitis. Collectively, our results show that the altered corneal biology of uninfected NK1R-/- mice along with an enhanced immunological response after ocular HSV-1 infection causes an early development of herpes stromal keratitis in NK1R-/- mice.


Subject(s)
Cornea/immunology , Cornea/pathology , Herpesvirus 1, Human/immunology , Keratitis, Herpetic/virology , Receptors, Neurokinin-1/physiology , Animals , CD4-Positive T-Lymphocytes/immunology , Conjunctiva/immunology , Conjunctiva/pathology , Conjunctiva/virology , Cornea/virology , Dendritic Cells/immunology , Herpesvirus 1, Human/physiology , Homeostasis , Interferon-gamma/immunology , Keratitis, Herpetic/immunology , Keratitis, Herpetic/physiopathology , Lymph Nodes/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neutrophils/immunology , Receptors, Neurokinin-1/deficiency , Receptors, Neurokinin-1/immunology , Viral Load
20.
Sci Rep ; 6: 32707, 2016 09 07.
Article in English | MEDLINE | ID: mdl-27599866

ABSTRACT

Breathing is generated by a respiratory network in the brainstem. At its core, a population of neurons expressing neurokinin-1 receptors (NK1R) and the peptide somatostatin (SST) form the preBötzinger Complex (preBötC), a site essential for the generation of breathing. PreBötC interneurons generate rhythm and follower neurons shape motor outputs by activating upper airway respiratory muscles. Since NK1R-expressing preBötC neurons are preferentially inhibited by µ-opioid receptors via activation of GIRK channels, NK1R stimulation may also involve GIRK channels. Hence, we identify the contribution of GIRK channels to rhythm, motor output and respiratory modulation by NK1Rs and SST. In adult rats, GIRK channels were identified in NK1R-expressing preBötC cells. Their activation decreased breathing rate and genioglossus muscle activity, an important upper airway muscle. NK1R activation increased rhythmic breathing and genioglossus muscle activity in wild-type mice, but not in mice lacking GIRK2 subunits (GIRK2(-/-)). Conversely, SST decreased rhythmic breathing via SST2 receptors, reduced genioglossus muscle activity likely through SST4 receptors, but did not involve GIRK channels. In summary, NK1R stimulation of rhythm and motor output involved GIRK channels, whereas SST inhibited rhythm and motor output via two SST receptor subtypes, therefore revealing separate circuits mediating rhythm and motor output.


Subject(s)
Brain Stem/physiology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/physiology , Receptors, Neurokinin-1/physiology , Somatostatin/physiology , Animals , Immunohistochemistry , Male , Mice , Rats , Rats, Wistar , Respiration
SELECTION OF CITATIONS
SEARCH DETAIL
...