Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 164
Filter
1.
Nat Commun ; 12(1): 2496, 2021 05 03.
Article in English | MEDLINE | ID: mdl-33941789

ABSTRACT

Memory formation is key for brain functioning. Uncovering the memory mechanisms is helping us to better understand neural processes in health and disease. Moreover, more specific treatments for fear-related disorders such as posttraumatic stress disorder and phobias may help to decrease their negative impact on mental health. In this line, the Tachykinin 2 (Tac2) pathway in the central amygdala (CeA) has been shown to be sufficient and necessary for the modulation of fear memory consolidation. CeA-Tac2 antagonism and its pharmacogenetic temporal inhibition impair fear memory in male mice. Surprisingly, we demonstrate here the opposite effect of Tac2 blockade on enhancing fear memory consolidation in females. Furthermore, we show that CeA-testosterone in males, CeA-estradiol in females and Akt/GSK3ß/ß-Catenin signaling both mediate the opposite-sex differential Tac2 pathway regulation of fear memory.


Subject(s)
Central Amygdaloid Nucleus/physiology , Conditioning, Classical/physiology , Fear/physiology , Memory Consolidation/physiology , Protein Precursors/antagonists & inhibitors , Tachykinins/antagonists & inhibitors , Animals , Antipsychotic Agents/pharmacology , Estradiol/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Piperidines/pharmacology , Protein Precursors/metabolism , Sex Factors , Signal Transduction , Tachykinins/metabolism , Testosterone/metabolism
2.
J Neurosci ; 41(5): 901-910, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33472824

ABSTRACT

Post-traumatic stress disorder (PTSD) is characterized by hypervigilance, increased reactivity to unpredictable versus predictable threat signals, deficits in fear extinction, and an inability to discriminate between threat and safety. First-line pharmacotherapies for psychiatric disorders have limited therapeutic efficacy in PTSD. However, recent studies have advanced our understanding of the roles of several limbic neuropeptides in the regulation of defensive behaviors and in the neural processes that are disrupted in PTSD. For example, preclinical studies have shown that blockers of tachykinin pathways, such as the Tac2 pathway, attenuate fear memory consolidation in mice and thus might have unique potential as early post-trauma interventions to prevent PTSD development. Targeting this pathway might also be beneficial in regulating other symptoms of PTSD, including trauma-induced aggressive behavior. In addition, preclinical and clinical studies have shown the important role of angiotensin receptors in fear extinction and the promise of using angiotensin II receptor blockade to reduce PTSD symptom severity. Additional preclinical studies have demonstrated that the oxytocin receptors foster accurate fear discrimination by facilitating fear responses to predictable versus unpredictable threats. Complementary human imaging studies demonstrate unique neural targets of intranasal oxytocin and compare its efficacy with well-established anxiolytic treatments. Finally, promising data from human subjects have demonstrated that a selective vasopressin 1A receptor antagonist reduces anxiety induced by unpredictable threats. This review highlights these novel promising targets for the treatment of unique core elements of PTSD pathophysiology.


Subject(s)
Anxiety/metabolism , Emotions/physiology , Limbic System/metabolism , Neuropeptides/metabolism , Stress Disorders, Post-Traumatic/metabolism , Animals , Anxiety/drug therapy , Anxiety/psychology , Emotions/drug effects , Extinction, Psychological/drug effects , Extinction, Psychological/physiology , Humans , Limbic System/drug effects , Nerve Net/drug effects , Nerve Net/metabolism , Neuropeptides/pharmacology , Neuropeptides/therapeutic use , Receptors, Tachykinin/antagonists & inhibitors , Receptors, Tachykinin/metabolism , Stress Disorders, Post-Traumatic/drug therapy , Stress Disorders, Post-Traumatic/psychology , Tachykinins/antagonists & inhibitors , Tachykinins/metabolism
3.
Mol Cancer Ther ; 18(12): 2368-2380, 2019 12.
Article in English | MEDLINE | ID: mdl-31439712

ABSTRACT

KRAS, an oncogene mutated in nearly one third of human cancers, remains a pharmacologic challenge for direct inhibition except for recent advances in selective inhibitors targeting the G12C variant. Here, we report that selective inhibition of the protein tyrosine phosphatase, SHP2, can impair the proliferation of KRAS-mutant cancer cells in vitro and in vivo using cell line xenografts and primary human tumors. In vitro, sensitivity of KRAS-mutant cells toward the allosteric SHP2 inhibitor, SHP099, is not apparent when cells are grown on plastic in 2D monolayer, but is revealed when cells are grown as 3D multicellular spheroids. This antitumor activity is also observed in vivo in mouse models. Interrogation of the MAPK pathway in SHP099-treated KRAS-mutant cancer models demonstrated similar modulation of p-ERK and DUSP6 transcripts in 2D, 3D, and in vivo, suggesting a MAPK pathway-dependent mechanism and possible non-MAPK pathway-dependent mechanisms in tumor cells or tumor microenvironment for the in vivo efficacy. For the KRASG12C MIAPaCa-2 model, we demonstrate that the efficacy is cancer cell intrinsic as there is minimal antiangiogenic activity by SHP099, and the effects of SHP099 is recapitulated by genetic depletion of SHP2 in cancer cells. Furthermore, we demonstrate that SHP099 efficacy in KRAS-mutant models can be recapitulated with RTK inhibitors, suggesting RTK activity is responsible for the SHP2 activation. Taken together, these data reveal that many KRAS-mutant cancers depend on upstream signaling from RTK and SHP2, and provide a new therapeutic framework for treating KRAS-mutant cancers with SHP2 inhibitors.


Subject(s)
Neoplasms/drug therapy , Neoplasms/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/genetics , Tachykinins/antagonists & inhibitors , Animals , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Mice , Neoplasms/pathology , Signal Transduction , Xenograft Model Antitumor Assays
4.
Cell ; 173(5): 1265-1279.e19, 2018 05 17.
Article in English | MEDLINE | ID: mdl-29775595

ABSTRACT

Chronic social isolation causes severe psychological effects in humans, but their neural bases remain poorly understood. 2 weeks (but not 24 hr) of social isolation stress (SIS) caused multiple behavioral changes in mice and induced brain-wide upregulation of the neuropeptide tachykinin 2 (Tac2)/neurokinin B (NkB). Systemic administration of an Nk3R antagonist prevented virtually all of the behavioral effects of chronic SIS. Conversely, enhancing NkB expression and release phenocopied SIS in group-housed mice, promoting aggression and converting stimulus-locked defensive behaviors to persistent responses. Multiplexed analysis of Tac2/NkB function in multiple brain areas revealed dissociable, region-specific requirements for both the peptide and its receptor in different SIS-induced behavioral changes. Thus, Tac2 coordinates a pleiotropic brain state caused by SIS via a distributed mode of action. These data reveal the profound effects of prolonged social isolation on brain chemistry and function and suggest potential new therapeutic applications for Nk3R antagonists.


Subject(s)
Brain/metabolism , Neurokinin B/metabolism , Protein Precursors/metabolism , Social Isolation , Stress, Psychological , Tachykinins/metabolism , Animals , Antipsychotic Agents/pharmacology , Behavior, Animal/drug effects , Brain/pathology , Female , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neurokinin B/genetics , Neurons/cytology , Neurons/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Precursors/antagonists & inhibitors , Protein Precursors/genetics , RNA Interference , RNA, Small Interfering/genetics , Receptors, Tachykinin/antagonists & inhibitors , Receptors, Tachykinin/metabolism , Tachykinins/antagonists & inhibitors , Tachykinins/genetics , Up-Regulation/drug effects
5.
Can J Physiol Pharmacol ; 94(9): 955-60, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27285300

ABSTRACT

The present study focused on the interactive pain regulation of endokinin A/B (EKA/B, the common C-terminal decapeptide in EKA and EKB) or endokinin C/D (EKC/D, the common C-terminal duodecapeptide in EKC and EKD) on chimeric peptide MCRT (YPFPFRTic-NH2, based on YPFP-NH2 and PFRTic-NH2) at the supraspinal level in mice. Results demonstrated that the co-injection of nanomolar EKA/B and MCRT showed moderate regulation, whereas 30 pmol EKA/B had no effect on MCRT. The combination of EKC/D and MCRT produced enhanced antinociception, which was nearly equal to the sum of the mathematical values of single EKC/D and MCRT. Mechanism studies revealed that pre-injected naloxone attenuated the combination significantly compared with the equivalent analgesic effects of EKC/D alone, suggesting that EKC/D and MCRT might act on two totally independent pathways. Moreover, based on the above results and previous reports, we made two reasonable hypotheses to explain the cocktail-induced analgesia, which may potentially pave the way to explore the respective regulatory mechanisms of EKA/B, EKC/D, and MCRT and to better understand the complicated pain regulation of NK1 and µ opioid receptors, as follows: (1) MCRT and endomorphin-1 possibly activated different µ subtypes; and (2) picomolar EKA/B might motivate the endogenous NPFF system after NK1 activation.


Subject(s)
Endorphins/pharmacology , Pain Measurement/drug effects , Peptide Fragments/pharmacology , Protein Precursors/pharmacology , Tachykinins/pharmacology , Analgesics/administration & dosage , Analgesics/pharmacology , Animals , Dose-Response Relationship, Drug , Drug Synergism , Endorphins/administration & dosage , Endorphins/antagonists & inhibitors , Infusions, Intraventricular , Male , Mice , Naloxone/pharmacology , Peptide Fragments/administration & dosage , Protein Precursors/administration & dosage , Protein Precursors/antagonists & inhibitors , Tachykinins/administration & dosage , Tachykinins/antagonists & inhibitors
7.
Curr Pharm Des ; 21(26): 3807-12, 2015.
Article in English | MEDLINE | ID: mdl-26044977

ABSTRACT

There is a wealth of evidence that various neuropeptides and their receptor ligands modulate schizophrenia- related behaviors in preclinical animal models, suggesting that neuropeptide systems may represent potential novel therapeutic targets for the treatment of schizophrenia. In particular, neurotensin and tachykinins have been the subject of significant research efforts, generating compelling preclinical data in the schizophrenia field. However, clinical studies with notably selective tachykinin NK3 receptor antagonists in schizophrenia have been disappointing, and they were unable to confirm the promising therapeutic potential from animal studies, thereby questioning the therapeutic utility of these compounds for this condition. This article reviews preclinical and clinical findings on ligands for neurotensin and tachykinin receptors in schizophrenia, and provides possible explanations for the failure so far to develop small-molecule neuropeptide ligands for the treatment of schizophrenia.


Subject(s)
Antipsychotic Agents/therapeutic use , Receptors, Neurotensin/antagonists & inhibitors , Receptors, Tachykinin/antagonists & inhibitors , Schizophrenia/drug therapy , Animals , Antipsychotic Agents/pharmacology , Humans , Ligands , Neuropeptides , Neurotensin/antagonists & inhibitors , Neurotensin/physiology , Receptors, Neurotensin/physiology , Receptors, Tachykinin/physiology , Schizophrenia/physiopathology , Tachykinins/antagonists & inhibitors , Tachykinins/physiology
8.
Proc Natl Acad Sci U S A ; 110(37): E3526-34, 2013 Sep 10.
Article in English | MEDLINE | ID: mdl-23980168

ABSTRACT

An arthropod-specific peptidergic system, the neuropeptide designated here as natalisin and its receptor, was identified and investigated in three holometabolous insect species: Drosophila melanogaster, Tribolium castaneum, and Bombyx mori. In all three species, natalisin expression was observed in 3-4 pairs of the brain neurons: the anterior dorso-lateral interneurons, inferior contralateral interneurons, and small pars intercerebralis neurons. In B. mori, natalisin also was expressed in two additional pairs of contralateral interneurons in the subesophageal ganglion. Natalisin-RNAi and the activation or silencing of the neural activities in the natalisin-specific cells in D. melanogaster induced significant defects in the mating behaviors of both males and females. Knockdown of natalisin expression in T. castaneum resulted in significant reduction in the fecundity. The similarity of the natalisin C-terminal motifs to those of vertebrate tachykinins and of tachykinin-related peptides in arthropods led us to identify the natalisin receptor. A G protein-coupled receptor, previously known as tachykinin receptor 86C (also known as the neurokinin K receptor of D. melanogaster), now has been recognized as a bona fide natalisin receptor. Taken together, the taxonomic distribution pattern of the natalisin gene and the phylogeny of the receptor suggest that natalisin is an ancestral sibling of tachykinin that evolved only in the arthropod lineage.


Subject(s)
Drosophila Proteins/physiology , Fertility/physiology , Insect Proteins/physiology , Insecta/physiology , Neuropeptides/physiology , Sexual Behavior, Animal/physiology , Tachykinins/physiology , Amino Acid Sequence , Animals , Bombyx/genetics , Bombyx/physiology , Brain/cytology , Brain/metabolism , Conserved Sequence , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Female , Fertility/genetics , Insect Proteins/antagonists & inhibitors , Insect Proteins/genetics , Insecta/genetics , Interneurons/metabolism , Male , Molecular Sequence Data , Neuropeptides/antagonists & inhibitors , Neuropeptides/genetics , Phylogeny , RNA Interference , Receptors, Tachykinin/genetics , Receptors, Tachykinin/physiology , Signal Transduction , Tachykinins/antagonists & inhibitors , Tachykinins/genetics , Tribolium/genetics , Tribolium/physiology
9.
Folia Neuropathol ; 51(2): 132-9, 2013.
Article in English | MEDLINE | ID: mdl-23821385

ABSTRACT

Opiate analgesics like morphine or fentanyl are the most widely used medicines for relieving severe acute or chronic pain, including cancer pain. Unfortunately, chronic pain treatment is associated with fast development of tolerance that creates the need to escalate the treatment doses. In addition, opiates may stimulate progression of cancer. Therefore, a new type of effective analgesic especially designed for chronic cancer pain treatment is needed. In this paper, a new opioid peptide analogue has been described as a new analgesic. The compound is characterized by very high agonist affinities to MOR and also high, but ten times lower affinity to DOR. Affinity to hNK1 as an antagonist is on the level of C-terminal hexapeptide fragment analogue of Substance P. The compound expressed reasonable antiproliferative properties toward various cancer cells. Interestingly, the peptide did not interfere with the proliferation of fibro-blasts. Therefore, the compound should be considered as a new analgesic for treatment of cancer-related pains with adjuvant anticancer properties which may support cancer treatments.


Subject(s)
Analgesics, Opioid/pharmacology , Antineoplastic Agents/pharmacology , Narcotic Antagonists/pharmacology , Receptors, Opioid/agonists , Tachykinins/antagonists & inhibitors , Adjuvants, Pharmaceutic/chemical synthesis , Adjuvants, Pharmaceutic/metabolism , Adjuvants, Pharmaceutic/pharmacology , Analgesics, Opioid/chemical synthesis , Analgesics, Opioid/metabolism , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , CHO Cells , Cell Line, Tumor , Cell Proliferation/drug effects , Chemotherapy, Adjuvant/methods , Cricetinae , Cricetulus , Humans , Narcotic Antagonists/chemical synthesis , Narcotic Antagonists/metabolism , Protein Binding/physiology , Rats, Wistar , Receptors, Neurokinin-1/physiology , Receptors, Opioid/metabolism , Tachykinins/physiology
10.
Peptides ; 43: 113-20, 2013 May.
Article in English | MEDLINE | ID: mdl-23470255

ABSTRACT

Hemokinin-1 is a peptide encoded by Pptc, which belongs to the family of mammalian tachykinins. Our previous results showed that rat/mouse hemokinin-1 (r/m HK-1) produced striking analgesia after intracerebroventricular (i.c.v.) injection in mice, and the analgesia could be blocked by the NK1 receptor antagonist and the opioid receptor antagonist, respectively. However, the precise distribution sites and the molecular mechanism involved in the analgesic effect after i.c.v. administration of r/m HK-1 are needed to be further investigated deeply. Using the fluorescence labeling method, our present results directly showed that r/m HK-1 peptides were mainly distributed at the ventricular walls and several juxta-ventricular structures for the first time. Our results showed that the mRNA expressions of NK1 receptor, PPT-A, PPT-C, KOR, PDYN, DOR and PENK were not changed markedly, as well as the protein expression of NK1 receptor was hardly changed. However, both the transcripts and proteins of MOR and POMC were up-regulated significantly, indicating that the analgesic effect induced by i.c.v. administration of r/m HK-1 is related to the activation of NK1 receptor first, then it is related to the release of endogenous proopiomelanocortin, as well as the increased expression level of µ opioid receptor. These results should facilitate further the analysis of the analgesia of r/m HK-1 in the central nerval system in acute pain and may open novel pharmacological interventions.


Subject(s)
Analgesia , Tachykinins/pharmacology , Tachykinins/pharmacokinetics , Animals , Female , Infusions, Intraventricular , Male , Mice , Mice, Inbred ICR , Narcotic Antagonists/pharmacology , Neurokinin-1 Receptor Antagonists/pharmacology , Opioid Peptides/genetics , Opioid Peptides/metabolism , Pro-Opiomelanocortin/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Neurokinin-1/genetics , Receptors, Neurokinin-1/metabolism , Receptors, Opioid/genetics , Receptors, Opioid/metabolism , Tachykinins/administration & dosage , Tachykinins/antagonists & inhibitors
11.
J Pharmacol Exp Ther ; 340(1): 27-36, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21957267

ABSTRACT

Hemokinin-1 (HK-1) is a newly identified tachykinin, originating from the immune system rather than neurons, and may participate in the immune and inflammatory response. In colonic mucosa of patients with inflammatory bowel disease (IBD), up-regulation of the TAC4 gene encoding HK-1 and increased production of prostaglandin E2 (PGE2) occur. Our aim was to examine the mechanistic link between human HK-1 and PGE2 production in normal human colon. Exogenous HK-1 (0.1 µM) for 4 h evoked an increased PGE2 release from colonic mucosal and muscle explants by 10- and 3.5-fold, respectively, compared with unstimulated time controls. The HK-1-stimulated PGE2 release was inhibited by the tachykinin receptor antagonists (S)1-2-[3-(3,4-dichlorophenyl)-1-(3-isopropoxyphenylacetyl)piperidin-3-yl]ethyl-4-phenyl-l azonia-bicyclo[2.2.2]octane (SR140333) [neurokinin-1 (NK1)] and N-[(2S)-4-(4-acetamido-4-phenylpiperidin-1-yl)-2-(3,4-dichlorophenyl)butyl]-N-methylbenzamide (SR48968) [neurokinin-2 (NK2)] and was also inhibited by the cyclooxygenase (COX)-2 inhibitor N-(2-cyclohexyloxy-4-nitrophenyl)methanesulfonamide) (NS-398) but not by the COX-1 inhibitor 5-(4-chlorophenyl)-1-(4-methoxyphenyl)-3-trifluoromethylpyrazole (SC-560). A parallel study with substance P showed similar results. Molecular studies with HK-1-treated explants demonstrated a stimulatory effect on COX-2 expression at both transcription and protein levels. It is noteworthy that this was coupled with HK-1-induced down-regulation of 15-hydroxyprostaglandin dehydrogenase (15-PGDH) mRNA and protein expression. Immunoreactivity for 15-PGDH occurred on inflammatory cells, epithelial cells, platelets, and ganglia. This finding provides an additional mechanism for HK-1-evoked PGE2 increase, in which HK-1 may interfere with the downstream metabolism of PGE2 by suppressing 15-PGDH expression. In conclusion, our results uncover a novel inflammatory role for HK-1, which signals via NK1 and NK2 receptors to regulate PGE2 release from human colonic tissue, and may further explain a pathological role for HK-1 in IBD when abnormal levels of PGE2 occur.


Subject(s)
Colon/metabolism , Cyclooxygenase 2/metabolism , Dinoprostone/biosynthesis , Enzyme Inhibitors/pharmacology , Hydroxyprostaglandin Dehydrogenases/antagonists & inhibitors , Tachykinins/pharmacology , Adult , Aged , Blotting, Western , Colitis/physiopathology , Colon/drug effects , Colon/enzymology , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression Regulation, Enzymologic/drug effects , Humans , Immunohistochemistry , In Vitro Techniques , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Male , Middle Aged , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Real-Time Polymerase Chain Reaction , Receptors, Neurokinin-1/drug effects , Receptors, Neurokinin-1/physiology , Receptors, Neurokinin-2/drug effects , Receptors, Neurokinin-2/physiology , Stimulation, Chemical , Tachykinins/antagonists & inhibitors
12.
Drug Metab Dispos ; 40(3): 419-25, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22184457

ABSTRACT

This article describes the combination of whole-body autoradiography with liquid extraction surface analysis (LESA) and mass spectrometry (MS) to study the distribution of the tachykinin neurokinin-1 antagonist figopitant and its metabolites in tissue sections of rats after intravenous administration of 5.0 mg/kg figopitant. An overview of autoradiography results is presented together with mass spectrometry identification and semiquantification of parent drug and its metabolites based on LESA-MS. The quality and accuracy of data generated by LESA-MS were assessed in comparison with classic tissue extraction, sample cleanup, and high-performance liquid chromatography analysis. The parent drug and the N-dealkylated metabolite M474(1) (BIIF 1148) in varying ratios were the predominant compounds in all tissues investigated. In addition, several metabolites formed by oxygenation, dealkylation, and a combination of oxygenation and dealkylation were identified. In summary, the LESA-MS technique was shown to be a powerful tool for identification and semiquantification of figopitant and its metabolites in different tissues and was complementary to quantitative whole-body autoradiography for studying the distribution.


Subject(s)
Antiemetics/pharmacokinetics , Mass Spectrometry/methods , Animals , Antiemetics/metabolism , Autoradiography/methods , Benzeneacetamides/metabolism , Benzeneacetamides/pharmacokinetics , Carbon Radioisotopes , Chromatography, High Pressure Liquid/methods , Liquid-Liquid Extraction/methods , Male , Neurokinin-1 Receptor Antagonists , Piperazines/metabolism , Piperazines/pharmacokinetics , Rats , Rats, Wistar , Tachykinins/antagonists & inhibitors , Tissue Distribution
13.
Peptides ; 31(8): 1613-6, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20451571

ABSTRACT

Hemokinin-1 is a novel mammalian tachykinin cloned from mouse bone marrow. At present, pharmacological profile and physiological role of hemokinin-1 are still unclear. In the present study, we found that intrathecal (i.t.) administration of hemokinin-1 (0.00625-1.6 nmol) induced nociceptive responses consisting of scratching, biting and licking, which resemble substance P-induced behavioral responses in mice. The behaviors evoked by low-dose of hemokinin-1 (0.0125 nmol) were dose-dependently inhibited by i.t. co-administration of CP-99,994, a non-peptidic tachykinin NK(1) receptor antagonist, whereas high-dose of hemokinin-1 (0.1 nmol)-induced behaviors were not affected. Moreover, sendide, a peptidic tachykinin NK(1) receptor antagonist, failed to reduce the behavioral responses of both low- and high-dose of hemokinin-1. In contrast, substance P-induced behaviors were completely suppressed by both CP-99,994 and sendide. These results suggest that hemokinin-1 plays an important role in pain transmission at spinal cord. Moreover, the mechanism of hemokinin-1-induced nociceptive behaviors may be dose-dependent, and distinct from substance P-induced nociceptive behaviors.


Subject(s)
Behavior, Animal/drug effects , Lumbar Vertebrae/innervation , Pain/physiopathology , Spinal Nerves/physiopathology , Synaptic Transmission/drug effects , Tachykinins/administration & dosage , Tachykinins/physiology , Analgesics/administration & dosage , Analgesics/therapeutic use , Animals , Dose-Response Relationship, Drug , Injections, Spinal , Male , Mice , Neurokinin-1 Receptor Antagonists , Neurotransmitter Agents/administration & dosage , Neurotransmitter Agents/therapeutic use , Pain/chemically induced , Pain/drug therapy , Pain Measurement , Peptide Fragments/administration & dosage , Peptide Fragments/therapeutic use , Piperidines/administration & dosage , Piperidines/therapeutic use , Pyrrolidonecarboxylic Acid/administration & dosage , Pyrrolidonecarboxylic Acid/analogs & derivatives , Pyrrolidonecarboxylic Acid/therapeutic use , Receptors, Neurokinin-1/metabolism , Spinal Nerves/drug effects , Substance P/administration & dosage , Substance P/antagonists & inhibitors , Substance P/physiology , Substance P/therapeutic use , Tachykinins/antagonists & inhibitors , Time Factors
14.
J Neurochem ; 103(6): 2153-63, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17949415

ABSTRACT

The tachykinin neurokinin 1 receptors (NK(1)Rs) regulation of acetylcholine release and its interaction with the enkephalin/mu opioid receptors (MORs) transmission was investigated in the limbic/prefrontal (PF) territory of the dorsal striatum. Using double immunohistochemistry, we first showed that in this territory, cholinergic interneurons contain tachykinin NK(1)Rs and co-express MORs in the last part of the light period (afternoon). In slices of the striatal limbic/PF territory, following suppression of the dopaminergic inhibitory control of acetylcholine release, application of the tachykinin NK(1)R antagonist, SSR240600, markedly reduced the NMDA-induced acetylcholine release in the morning but not in the afternoon when the enkephalin/MOR regulation is operational. In the afternoon, the NK(1)R antagonist response required the suppression of the enkephalin/MOR inhibitory control of acetylcholine release by betafunaltrexamine. The pharmacological profile of the tachykinin NK(1)R regulation tested by application of the receptor agonists [[Pro(9)]substance P, neurokinin A, neuropeptide K, and substance P(6-11)] and antagonists (SSR240600, GR205171, GR82334, and RP67580) indicated that the subtype of tachykinin NK(1)R implicated are the new NK(1)-sensitive receptor binding site. Therefore, in the limbic/PF territory of the dorsal striatum, endogenous tachykinin facilitates acetylcholine release via a tachykinin NK(1)R subtype. In the afternoon, the tachykinin/NK(1)R and the enkephalin/MOR transmissions interact to control cholinergic transmission.


Subject(s)
Cholinergic Fibers/metabolism , Neostriatum/metabolism , Receptors, Neurokinin-1/metabolism , Receptors, Opioid, mu/metabolism , Synaptic Transmission/physiology , Tachykinins/metabolism , Acetylcholine/metabolism , Animals , Binding Sites/drug effects , Binding Sites/physiology , Binding, Competitive/drug effects , Binding, Competitive/physiology , Circadian Rhythm/drug effects , Circadian Rhythm/physiology , Enkephalins/metabolism , Limbic System/metabolism , Male , Morpholines/pharmacology , Narcotic Antagonists/pharmacology , Neostriatum/cytology , Neural Pathways/metabolism , Neurokinin-1 Receptor Antagonists , Organ Culture Techniques , Piperidines/pharmacology , Prefrontal Cortex/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Neurokinin-1/agonists , Receptors, Opioid, mu/antagonists & inhibitors , Synaptic Transmission/drug effects , Tachykinins/agonists , Tachykinins/antagonists & inhibitors
15.
Biochem Pharmacol ; 74(5): 752-7, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17632084

ABSTRACT

Topically applied morphine is routinely used to alleviate pain in cutaneous wounds such as burns and pressure sores. Evidence suggests the topical administration of exogenous opioid drugs may impair wound closure. This study examined the effects of topical morphine on a standardized model of cutaneous wound healing in the rat. Full-thickness 4mm diameter circular skin flaps were excised from the intrascapular region of male Sprague-Dawley rats. IntraSite Gel infused with either morphine-sulfate, neurokinin-1 (NK-1) or neurokinin-2 (NK-2) receptor antagonists, substance P (SP), neurokinin A (NKA), SP+morphine-sulfate, or NKA+morphine-sulfate was applied to the wound twice daily. Results demonstrated a significant overall delay in the time course of wound contraction in morphine-treated animals when compared with gel-only treated controls. The delay in wound contraction seen in morphine-treated animals increased in a concentration-dependent manner. Topical application of NK-1 or NK-2 receptor antagonists mimicked the effects of morphine in delaying wound closure, suggesting topical opioids impair wound closure via the inhibition of SP and NKA release peripherally into the healing wound. Additionally, no significant delays in closure were seen in rats receiving morphine combined with SP or NKA, demonstrating the ability of each neuropeptide to attenuate the effects of morphine in delaying wound closure and restore normal wound closure rates. The combination of SP or NKA and morphine-sulfate for wound therapy may provide local analgesia while maintaining normal closure rates.


Subject(s)
Morphine/administration & dosage , Morphine/adverse effects , Tachykinins/antagonists & inhibitors , Wound Healing/drug effects , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/adverse effects , Analgesics, Opioid/pharmacology , Animals , Dose-Response Relationship, Drug , Indoles/administration & dosage , Indoles/adverse effects , Indoles/pharmacology , Male , Morphine/pharmacology , Pain/drug therapy , Piperidines/administration & dosage , Piperidines/adverse effects , Piperidines/pharmacology , Rats , Rats, Sprague-Dawley
17.
Infect Immun ; 75(4): 1895-903, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17261611

ABSTRACT

Bacillus anthracis edema toxin (ET), composed of protective antigen and an adenylate cyclase edema factor (EF), elicits edema in host tissues, but the target cells and events leading from EF-mediated cyclic-AMP production to edema are unknown. We evaluated the direct effect of ET on several cell types in vitro and tested the possibility that mediators of vascular leakage, such as histamine, contribute to edema in rabbits given intradermal ET. ET increased the transendothelial electrical resistance of endothelial monolayers, a response that is mechanistically inconsistent with the in vivo vascular leakage induced by ET. Screening of several drugs by intradermal treatment prior to toxin injection demonstrated reduced ET-induced vascular leakage with a cyclo-oxygenase inhibitor (indomethacin), agents that interfere with histamine (pyrilamine or cromolyn), or a neurokinin antagonist (spantide). Systemic administration of indomethacin or celecoxib (cyclo-oxygenase inhibitors), pyrilamine, aprepitant (a neurokinin 1 receptor antagonist), or indomethacin with pyrilamine significantly reduced vascular leakage associated with ET. Although the effects of pyrilamine, cromolyn, or aprepitant on ET-induced vascular leakage suggest a possible role for mast cells (MC) and sensory neurons in ET-induced edema, ET did not elicit degranulation of human skin MC or substance P release from NT2N cells in vitro. Our results indicate that ET, acting indirectly or directly on a target yet to be identified, stimulates the production/release of multiple inflammatory mediators, specifically neurokinins, prostanoids, and histamine. These mediators, individually and through complex interactions, increase vascular permeability, and interventions directed at these mediators may benefit hosts infected with B. anthracis.


Subject(s)
Antigens, Bacterial/toxicity , Bacillus anthracis , Bacterial Toxins/toxicity , Edema/chemically induced , Histamine/metabolism , Prostaglandins/metabolism , Tachykinins/metabolism , Animals , Aprepitant , Capillary Permeability/drug effects , Celecoxib , Cell Degranulation , Cell Line , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Histamine H1 Antagonists/pharmacology , Humans , Indomethacin/pharmacology , Injections, Intradermal , Mast Cells/drug effects , Morpholines/pharmacology , Neurokinin-1 Receptor Antagonists , Prostaglandin Antagonists/pharmacology , Pyrazoles/pharmacology , Pyrilamine/pharmacology , Rabbits , Substance P/analogs & derivatives , Substance P/metabolism , Substance P/pharmacology , Sulfonamides/pharmacology , Tachykinins/antagonists & inhibitors
18.
Curr Drug Targets ; 7(8): 1021-9, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16918330

ABSTRACT

Tachykinins (TKs) and their receptors (NK1, NK2 and NK3), which are diffusely expressed in the human gastrointestinal tract, represent an endogenous modulator system regulating enteric secretomotor functions, inflammatory and immune responses, and visceral hypersensitivity, mainly during pathological gut diseases. Pathophysiological implications of TKs in the digestive tract include changes in TK innervation, in the expression of TKs and TK receptors, which result in inflammation- and immune-induced disturbances of gut functions, such as dysmotility (diarrhoea/constipation), secretory diarrhoea and visceral hyperalgesia. Increasing evidence correlates all these TKergic system abnormalities with gastrointestinal diseases of different etiology (i.e. inflammatory bowel diseases, irritable bowel syndrome). Accordingly, TK receptors have been identified as novel targets for the development of new therapeutic agents for clinical use. Available preclinical findings have shown that TK antagonists could counteract the most significant symptoms characterizing these gut diseases.


Subject(s)
Gastrointestinal Tract/pathology , Gastrointestinal Tract/physiology , Receptors, Tachykinin/physiology , Tachykinins/physiology , Animals , Gastrointestinal Diseases/metabolism , Gastrointestinal Diseases/pathology , Gastrointestinal Tract/chemistry , Gastrointestinal Tract/metabolism , Humans , Receptors, Tachykinin/biosynthesis , Tachykinins/antagonists & inhibitors , Tachykinins/biosynthesis , Tachykinins/metabolism
19.
Auton Neurosci ; 126-127: 273-6, 2006 Jun 30.
Article in English | MEDLINE | ID: mdl-16567131

ABSTRACT

We investigated the participation of different tachykinin receptors in contractility of circular muscle strips of the mouse ileum using selective NK receptor agonists and antagonists. The NK1 receptor agonist septide (1-100 nM) induced dose-dependent contractions which were reduced by atropine and augmented by L-NNA. L-NNA increased and TTX consecutively reduced contractions to the NK2 receptor agonist beta-A-NKA (1-100 nM). Senktide, agonist of NK3 receptors, failed to induce contractions. NANC contractions to EFS were decreased after NK1 receptor blockade with RP67580. This inhibitory effect was more pronounced after additional blockade of NK2 and NK3 receptors. NK3 receptor antagonism alone reduced contractions at higher frequencies of stimulation. When the duration of the EFS stimulus was increased, the participation of all NK receptor subtypes became more evident. Our results suggest that excitatory NANC transmission in the circular muscle layer of the mouse ileum is mediated by tachykinins acting principally on NK1 receptors on cholinergic nerves and smooth muscle cells. Also NK2 receptors, located on smooth muscle cells and nitrergic neurons, and NK3 receptors on enteric neurons are involved.


Subject(s)
Muscle Contraction/physiology , Muscle, Smooth/blood supply , Muscle, Smooth/physiology , Tachykinins/physiology , Animals , Atropine/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Inhibitors/pharmacology , Ilium/cytology , In Vitro Techniques , Male , Mice , Muscarinic Antagonists/pharmacology , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Nitroarginine/pharmacology , Peptide Fragments/pharmacology , Potassium Chloride/pharmacology , Pyrrolidonecarboxylic Acid/analogs & derivatives , Pyrrolidonecarboxylic Acid/pharmacology , Substance P/analogs & derivatives , Substance P/pharmacology , Tachykinins/agonists , Tachykinins/antagonists & inhibitors , Tetrodotoxin/pharmacology
20.
Neuroscience ; 139(2): 495-503, 2006 May 12.
Article in English | MEDLINE | ID: mdl-16458437

ABSTRACT

The objective of the present study was to investigate the hypothesis of the presence of a local neural reflex modulating the vagally mediated contractions of striated muscle in the rat esophagus and to determine the possible involvement of tachykinins in such a local neural reflex. Electrical stimulation of the vagus nerve evoked twitch contractile responses that were abolished by d-tubocurarine (5 microM). Capsaicin (1-100 microM) inhibited the vagally mediated twitch contractions o f the normal rat esophageal preparations concentration-dependently but not those of the neonatally capsaicin-treated ones. NG-nitro-L-arginine methyl ester (100 microM), a nitric oxide synthase inhibitor, blocked the inhibitory effect of capsaicin and exogenous application of a nitric oxide donor (1 mM) inhibited the vagally mediated twitch contractions. Capsaicin suppressed acetylcholine release from the normal rat esophageal segments evoked by vagus nerve stimulation but not that from the neonatally capsaicin-treated ones. A selective tachykinin NK1 receptor antagonist (0.1 or 1 microM) attenuated the inhibitory effect of capsaicin. However, antagonists of tachykinin NK2, tachykinin NK3 and calcitonin gene-related peptide receptors (1 microM) did not have any effect. A tachykinin NK1 receptor agonist (1 or 5 microM) inhibited the vagally mediated twitch contractions, which was prevented by NG-nitro-L-arginine methyl ester (100 microM). These data suggest that the rat esophagus might have a local neural reflex inhibiting the vagally mediated striated muscle motility, which consists of capsaicin-sensitive sensory neurons and myenteric nitrergic neurons, and that tachykinins might be involved in the neural reflex through tachykinin NK1 receptors.


Subject(s)
Esophagus/cytology , Muscle Contraction/physiology , Muscle, Skeletal/physiology , Reflex/physiology , Tachykinins/physiology , Acetylcholine/metabolism , Animals , Animals, Newborn , Atropine/pharmacology , Capsaicin/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Inhibitors/pharmacology , In Vitro Techniques , Male , Muscarinic Antagonists/pharmacology , Muscle Contraction/radiation effects , Muscle, Skeletal/radiation effects , NG-Nitroarginine Methyl Ester/pharmacology , Neurotransmitter Agents/pharmacology , Nicotinic Antagonists/pharmacology , Nitric Oxide Donors/pharmacology , Physical Stimulation/methods , Rats , Rats, Wistar , Tachykinins/antagonists & inhibitors , Tubocurarine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...