Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 150
Filter
1.
Peptides ; 146: 170651, 2021 12.
Article in English | MEDLINE | ID: mdl-34560171

ABSTRACT

Orexin 1 receptors (Orx1R) and cannabinoid 1 receptors (CB1R) are implicated in migraine pathophysiology. This study evaluated the potential involvement of Orx1R and CB1R within the ventrolateral periaqueductal gray matter (vlPAG) in the modulation of anxiety-like behavior and social interaction of migraineurs rats. A rat model of migraine induced by recurrent administration of nitroglycerin (NTG) (5 mg/kg/i.p.). The groups of rats (n = 6) were then subjected to intra-vlPAG microinjection of orexin-A (25, 50 pM), and Orx1R antagonist SB334867 (20, 40 nM) or AM 251 (2, 4 µg) as a CB1R antagonist. Behavioral responses were evaluated in elevated plus maze (EPM), open field (OF) and three-chambered social test apparatus. NTG produced a marked anxiety like behaviors, in both EPM and OF tasks. It did also decrease social performance. NTG-related anxiety and social conflicts were attenuated by orexin-A (25, 50 pM). However, NTG effects were exacerbated by SB334867 (40 nM) and AM251 (2, 4 µg). The orexin-A-mediated suppression of NTG-induced anxiety and social conflicts were prevented by either SB334867 (20 nM) or AM251 (2 µg). The findings suggest roles for Orx1R and CB1R signaling within vlPAG in the modulation of migraine-induced anxiety-like behavior and social dysfunction in rats.


Subject(s)
Anxiety/prevention & control , Behavior, Animal , Migraine Disorders/complications , Orexin Receptors/metabolism , Periaqueductal Gray/metabolism , Receptor, Cannabinoid, CB1/metabolism , Social Behavior , Animals , Anxiety/etiology , Benzoxazoles/pharmacology , Male , Naphthyridines/pharmacology , Piperidines/pharmacology , Pyrazoles/pharmacology , Rats , Rats, Wistar , Receptors, Neuropeptide/drug effects , Urea/analogs & derivatives , Urea/pharmacology
2.
Basic Clin Pharmacol Toxicol ; 128(6): 731-740, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33533572

ABSTRACT

Chimeric peptide MCRT (YPFPFRTic-NH2 ) was a multifunctional ligand of opioid and neuropeptide FF (NPFF) receptors and reported to be potentially antalgic in acute tail-flick test. Here, we developed spared nerve injury (SNI) model to explore its efficacy in chronic neuropathic pain. Analgesic tolerance, opioid-induced hyperalgesia and gastrointestinal transit were measured for safety evaluation. Intracerebroventricular (i.c.v.) and intraplantar (i.pl.) injections were conducted as central and peripheral routes, respectively. Results demonstrated that MCRT alleviated neuropathic pain effectively and efficiently, with the ED50 values of 4.93 nmol/kg at the central level and 3.11 nmol/kg at the peripheral level. The antagonist blocking study verified the involvement of mu-, delta-opioid and NPFF receptors in MCRT produced anti-allodynia. Moreover, the separation of analgesia from unwanted effects was preliminarily achieved and that MCRT caused neither analgesic tolerance nor hyperalgesia after chronic i.c.v. administration, nor constipation after i.pl. administration. Notably, the local efficacy of MCRT in SNI mice was about one thousandfold higher than morphine and ten thousandfold higher than pregabalin, indicating a great promise in the future treatment of neuropathic pain.


Subject(s)
Analgesics, Opioid/pharmacology , Endorphins/pharmacology , Neuralgia/drug therapy , Receptors, Neuropeptide/drug effects , Receptors, Opioid/drug effects , Animals , Ligands , Mice , Morphine , Narcotic Antagonists/pharmacology , Receptors, Opioid, mu/agonists
3.
Clin Exp Allergy ; 51(5): 685-695, 2021 05.
Article in English | MEDLINE | ID: mdl-33275825

ABSTRACT

BACKGROUND: Neuromuscular-blocking agents (NMBAs) can cause both IgE-dependent and IgE-independent anaphylactic reactions, with activation of the mast cell receptor MRGPRX2 being important to the latter. Sugammadex, a reversal agent for certain aminosteroid NMBAs, has been proposed as an antidote for these anaphylactic events with conflicting outcomes. OBJECTIVE: We further characterize the involvement of MRGPRX2 in NMBA-induced mast cell activation and determine how this is influenced by sugammadex. We then apply these in vitro results to infer the possible utility of sugammadex in the acute management of non-IgE-dependent anaphylaxis. METHODS: The LAD2 human mast cell line and a MRGPRX2 knock-down derivative were used to validate the involvement of MRGPRX2 and to test the effect of sugammadex on mast cell activation by NMBAs and other MRGPRX2 agonists. RESULTS: All MRGPRX2 agonists tested were shown to induce MRGPRX2-dependent LAD2 mast cell calcium mobilization and cytokine release and all, apart from rocuronium, induced degranulation. Co-treatment of mast cells with sugammadex and some MRGPRX2 agonists significantly reduced cell activation, but if sugammadex was administered a few minutes following stimulation, degranulation was not attenuated. However, addition of sugammadex up to 180 min following LAD2 MRGPRX2 stimulation, significantly reduced CCL2 mRNA and protein induction. CONCLUSIONS AND CLINICAL RELEVANCE: We show that sugammadex, known to reverse muscle blockade by certain NMBAs, is also able to reduce MRGPRX2 activation by NMBAs and other, but not all, MRGPRX2 agonists. As sugammadex was ineffective in attenuating mast cell degranulation when added rapidly post MRGPRX2 activation, this suggests against the agent having efficacy in controlling acute symptoms of anaphylaxis to NMBAs caused by MRGPRX2 activation. Interestingly, however, sugammadex did impair MRGPRX2-induced CCL2 release, suggesting that it may have some benefit in perhaps dampening less well-defined adverse effects of MRGPRX2-dependent anaphylaxis associated with the more slowly elaborated mast cell mediators.


Subject(s)
Anaphylaxis/drug therapy , Chemokine CCL2/drug effects , Mast Cells/drug effects , Nerve Tissue Proteins/drug effects , Neuromuscular Blocking Agents/pharmacology , Receptors, G-Protein-Coupled/drug effects , Receptors, Neuropeptide/drug effects , Sugammadex/pharmacology , Anaphylaxis/chemically induced , Antidotes/pharmacology , Atracurium/adverse effects , Cell Line , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Gene Knockdown Techniques , Humans , In Vitro Techniques , Mast Cells/immunology , Mast Cells/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuromuscular Blocking Agents/adverse effects , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism , Rocuronium/adverse effects
4.
Proc Natl Acad Sci U S A ; 115(47): 12028-12033, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30373845

ABSTRACT

The effects of the growth hormone-releasing hormone (GHRH) agonist MR409 on various human cancer cells were investigated. In H446 small cell lung cancer (SCLC) and HCC827 and H460 (non-SCLC) cells, MR409 promoted cell viability, reduced cell apoptosis, and induced the production of cellular cAMP in vitro. Western blot analyses showed that treatment of cancer cells with MR409 up-regulated the expression of cyclins D1 and D2 and cyclin-dependent kinases 4 and 6, down-regulated p27kip1, and significantly increased the expression of the pituitary-type GHRH receptor (pGHRH-R) and its splice-variant (SV1). Hence, in vitro MR409 exerts agonistic action on lung cancer cells in contrast to GHRH antagonists. However, in vivo, MR409 inhibited growth of lung cancers xenografted into nude mice. MR409 given s.c. at 5 µg/day for 4 to 8 weeks significantly suppressed growth of HCC827, H460, and H446 tumors by 48.2%, 48.7%, and 65.6%, respectively. This inhibition of tumor growth by MR409 was accompanied by the down-regulation of the expression of pGHRH-R and SV1 in the pituitary gland and tumors. Tumor inhibitory effects of MR409 in vivo were also observed in other human cancers, including gastric, pancreatic, urothelial, prostatic, mammary, and colorectal. This inhibition of tumor growth parallel to the down-regulation of GHRH-Rs is similar and comparable to the suppression of sex hormone-dependent cancers after the down-regulation of receptors for luteinizing hormone-releasing hormone (LHRH) by LHRH agonists. Further oncological investigations with GHRH agonists are needed to elucidate the underlying mechanisms.


Subject(s)
Receptors, Neuropeptide/drug effects , Receptors, Pituitary Hormone-Regulating Hormone/drug effects , Sermorelin/analogs & derivatives , Alternative Splicing/drug effects , Animals , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Down-Regulation/drug effects , Female , Growth Hormone-Releasing Hormone/agonists , Growth Hormone-Releasing Hormone/pharmacology , Humans , Mice , Mice, Nude , RNA Splicing/drug effects , Sermorelin/metabolism , Sermorelin/pharmacology , Small Cell Lung Carcinoma/metabolism , Xenograft Model Antitumor Assays/methods
5.
PLoS One ; 13(2): e0193058, 2018.
Article in English | MEDLINE | ID: mdl-29474388

ABSTRACT

Natalisins (NTLs) are conservative neuropeptides, which are only found in arthropods and are documented to regulate reproductive behaviors in insects. In our previous study, we have confirmed that NTLs regulate the reproductive process in an important agricultural pest, Bactrocera dorsalis (Hendel). Hence, in this study, to further confirm the in vivo function of NTL receptor (NTLR) and assess the potential of NTLR as an insecticide target, RNA interference targeting NTLR mRNA was performed. We found that mating frequencies of both males and females were reduced by RNAi-mediated knockdown of the NTLR transcript, while there was no effect on mating duration. Moreover, we functionally expressed the B. dorsalis NTLR in Chinese Hamster Ovary (CHO) cells and was co-transfected with an aequorin reporter to measure ligand activities. A total of 13 biostable multi-Aib analogs were tested for agonistic and antagonistic activities. While most of these NTL analogs did not show strong activity, one analog (NLFQV[Aib]DPFF[Aib]TRamide) had moderate antagonistic activity. Taken together, we provided evidence for the important roles of NTLR in regulating mating frequencies of both male and female in this fly and also provided in vitro data on mimetic analogs that serve as leading structures for the development of agonists and antagonists to disrupt the NTL signaling pathway.


Subject(s)
Insect Proteins/physiology , Neuropeptides/physiology , Peptidomimetics/pharmacology , Receptors, Neuropeptide/physiology , Sexual Behavior, Animal/physiology , Tephritidae/physiology , Amino Acid Sequence , Animals , CHO Cells , Cricetulus , Female , Gene Knockdown Techniques , Genes, Insect , Insect Proteins/drug effects , Insect Proteins/genetics , Male , Peptidomimetics/chemistry , Receptors, Neuropeptide/drug effects , Receptors, Neuropeptide/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Sexual Behavior, Animal/drug effects , Signal Transduction/drug effects , Tephritidae/genetics
6.
Neuropeptides ; 67: 36-44, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29195839

ABSTRACT

Neuropeptide S (NPS), the endogenous neuropeptide ligand of NPSR, has been reported to regulate anxiety-related behavior involved in multiple brain regions, including amygdale, locus coeruleus and Barrington's nucleus. However, little research has been conducted on the anxiolytic-like behaviors of NPS on the hypothalamus, which was an important area in defensive behavior. Here, we investigated a role of hypothalamus in anxiolytic-like behaviors of NPS. We found that NPSR protein of mouse distributed mainly in the ventromedial hypothalamus (VMH). And in the single prolonged stress model (SPS), the results showed that NPS mRNA of the mice exposed to SPS was significantly higher than control, while NPSR mRNA was remarkable lower than control in hypothalamus. Further studies found that NPS intra-VMH infusion dose-dependently (1, 10 and 100pmol) induced anxiolytic effects, using elevated plus maze and open field tests. These anxiolytic effects could be blocked by NPSR antagonist (SHA68), but not by picrotoxin (a GABAA receptor antagonist) and sacolfen (a GABAB receptor antagonist). Meanwhile, our data showed that the expression of c-Fos was significantly increased in VMH after NPS delivered into the lateral ventricles. These results cast a new light on the hypothalamic nucleus in the anxiolytic-like effect of NPS system.


Subject(s)
Anti-Anxiety Agents/pharmacology , Hypothalamus/drug effects , Motor Activity/drug effects , Neuropeptides/pharmacology , Receptors, Neuropeptide/metabolism , Animals , Male , Maze Learning/drug effects , Mice , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/metabolism , Receptors, Neuropeptide/drug effects
7.
J Neurosci ; 37(50): 12214-12225, 2017 12 13.
Article in English | MEDLINE | ID: mdl-29118105

ABSTRACT

Neuropeptides, such as neuropeptide S (NPS) and oxytocin (OXT), represent potential options for the treatment of anxiety disorders due to their potent anxiolytic profile. In this study, we aimed to reveal the mechanisms underlying the behavioral action of NPS, and present a chain of evidence that the effects of NPS within the hypothalamic paraventricular nucleus (PVN) are mediated via actions on local OXT neurons in male Wistar rats. First, retrograde studies identified NPS fibers originating in the brainstem locus coeruleus, and projecting to the PVN. FACS identified prominent NPS receptor expression in PVN-OXT neurons. Using genetically encoded calcium indicators, we further demonstrated that NPS reliably induces a transient increase in intracellular Ca2+ concentration in a subpopulation of OXT neurons, an effect mediated by NPS receptor. In addition, intracerebroventricular (i.c.v.) NPS evoked a significant somatodendritic release of OXT within the PVN as assessed by microdialysis in combination with a highly sensitive radioimmunoassay. Finally, we could show that the anxiolytic effect of NPS seen after i.c.v. or intra-PVN infusion requires responsive OXT neurons of the PVN and locally released OXT. Thus, pharmacological blockade of OXT receptors as well as chemogenetic silencing of OXT neurons within the PVN prevented the effect of synthetic NPS. In conclusion, our results indicate a significant role of the OXT system in mediating the effects of NPS on anxiety, and fill an important gap in our understanding of brain neuropeptide interactions in the context of regulation of emotional behavior within the hypothalamus.SIGNIFICANCE STATEMENT Given the rising scientific interest in neuropeptide research in the context of emotional and stress-related behaviors, our findings demonstrate a novel intrahypothalamic mechanism involving paraventricular oxytocin neurons that express the neuropeptide S receptor. These neurons respond with transient Ca2+ increase and somatodendritic oxytocin release following neuropeptide S stimulation. Thereby, oxytocin neurons seem essential for neuropeptide S-induced anxiolysis, as this effect was blocked by pharmacological and chemogenetic inhibition of the oxytocin system.


Subject(s)
Anxiety/physiopathology , Neuropeptides/physiology , Oxytocin/physiology , Paraventricular Hypothalamic Nucleus/physiology , Receptors, Oxytocin/physiology , Animals , Axonal Transport , Bacterial Proteins/analysis , Calcium Signaling/physiology , Dependovirus/genetics , Exploratory Behavior/drug effects , Genes, Reporter , Genetic Vectors , Luminescent Proteins/analysis , Male , Microdialysis , Motor Activity/drug effects , Neuropeptides/pharmacology , Oxytocin/agonists , Paraventricular Hypothalamic Nucleus/drug effects , Rats , Rats, Wistar , Receptors, Neuropeptide/drug effects , Receptors, Neuropeptide/physiology , Receptors, Oxytocin/antagonists & inhibitors , Synaptic Transmission/drug effects , Red Fluorescent Protein
8.
Neuropeptides ; 62: 81-86, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27993374

ABSTRACT

The amygdaloid body (AMY) plays an important role in memory, learning and reward-related processes. RFRP-1 immunoreactive fibers and NPFF receptors were identified in the AMY, and previously we verified that RFRP-1 infused into the central nucleus of AMY (CeA) induced place preference. The aim of the present study was to examine the possible effects of RFRP-1 in the CeA on passive avoidance learning. Male Wistar rats were examined in two-compartment passive avoidance paradigm. Animals were shocked with 0.5mA current and subsequently were microinjected bilaterally with 50ng or 100ng RFRP-1 in volume of 0.4µl, or 20ng NPFF receptor antagonist RF9 (ANT) alone, or antagonist 15min before 50ng RFRP-1 treatments into the CeA. Fifty nanogram dose of RFRP-1 significantly increased the step-through latency time, the 100ng RFRP-1 and the ANT alone were ineffective. The effect of 50ng RFRP-1 was eliminated by the ANT pretreatment. Our results suggest that intraamygdaloid RFRP-1 enhances learning processes and memory in aversive situations and this effect can specifically be prevented by ANT pretreatment.


Subject(s)
Avoidance Learning/drug effects , Central Amygdaloid Nucleus/drug effects , Memory/drug effects , Neuropeptides/pharmacology , Animals , Behavior, Animal/drug effects , Male , Microinjections/methods , Neuropeptides/metabolism , Rats, Wistar , Receptors, Neuropeptide/drug effects
9.
Nat Rev Gastroenterol Hepatol ; 13(1): 38-48, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26392067

ABSTRACT

The gastrointestinal tract is the major source of the related hormones ghrelin and motilin, which act on structurally similar G protein-coupled receptors. Nevertheless, selective receptor agonists are available. The primary roles of endogenous ghrelin and motilin in the digestive system are to increase appetite or hedonic eating (ghrelin) and initiate phase III of gastric migrating myoelectric complexes (motilin). Ghrelin and motilin also both inhibit nausea. In clinical trials, the motilin receptor agonist camicinal increased gastric emptying, but at lower doses reduced gastroparesis symptoms and improved appetite. Ghrelin receptor agonists have been trialled for the treatment of diabetic gastroparesis because of their ability to increase gastric emptying, but with mixed results; however, relamorelin, a ghrelin agonist, reduced nausea and vomiting in patients with this disorder. Treatment of postoperative ileus with a ghrelin receptor agonist proved unsuccessful. Centrally penetrant ghrelin receptor agonists stimulate defecation in animals and humans, although ghrelin itself does not seem to control colorectal function. Thus, the most promising uses of motilin receptor agonists are the treatment of gastroparesis or conditions with slow gastric emptying, and ghrelin receptor agonists hold potential for the reduction of nausea and vomiting, and the treatment of constipation. Therapeutic, gastrointestinal roles for receptor antagonists or inverse agonists have not been identified.


Subject(s)
Gastrointestinal Agents/therapeutic use , Gastrointestinal Diseases/drug therapy , Receptors, Gastrointestinal Hormone/drug effects , Receptors, Ghrelin/drug effects , Receptors, Neuropeptide/drug effects , Appetite/drug effects , Appetite/physiology , Constipation/drug therapy , Constipation/physiopathology , Gastric Emptying/drug effects , Gastric Emptying/physiology , Gastrointestinal Agents/adverse effects , Gastrointestinal Motility/drug effects , Gastrointestinal Motility/physiology , Ghrelin/physiology , Humans , Hunger/drug effects , Hunger/physiology , Motilin/physiology , Nausea/drug therapy , Nausea/physiopathology , Receptors, Gastrointestinal Hormone/agonists , Receptors, Gastrointestinal Hormone/physiology , Receptors, Ghrelin/agonists , Receptors, Ghrelin/physiology , Receptors, Neuropeptide/agonists , Receptors, Neuropeptide/physiology , Signal Transduction/physiology
10.
J Med Chem ; 58(22): 8834-49, 2015 Nov 25.
Article in English | MEDLINE | ID: mdl-26466164

ABSTRACT

Analogues of the argininamide-type NPY Y1 receptor (Y1R) antagonist BIBP3226, bearing carbamoyl moieties at the guanidine group, revealed subnanomolar Ki values and caused depression of the maximal response to NPY (calcium assay) by up to 90% in a concentration- and time-dependent manner, suggesting insurmountable antagonism. To gain insight into the mechanism of binding of the synthesized compounds, a tritiated antagonist, (R)-N(α)-diphenylacetyl-N(ω)-[2-([2,3-(3)H]propionylamino)ethyl]aminocarbonyl-(4-hydroxybenzyl)arginin-amide ([(3)H]UR-MK299, [(3)H]38), was prepared. [(3)H]38 revealed a dissociation constant in the picomolar range (Kd 0.044 nM, SK-N-MC cells) and very high Y1R selectivity. Apart from superior affinity, a considerably lower target off-rate (t1/2 95 min) was characteristic of [(3)H]38 compared to that of the higher homologue containing a tetramethylene instead of an ethylene spacer (t1/2 3 min, Kd 2.0 nM). Y1R binding of [(3)H]38 was fully reversible and fully displaceable by nonpeptide antagonists and the agonist pNPY. Therefore, the insurmountable antagonism observed in the functional assay has to be attributed to the extended target-residence time, a phenomenon of relevance in drug research beyond the NPY receptor field.


Subject(s)
Arginine/analogs & derivatives , Arginine/chemistry , Diphenylacetic Acids/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , Receptors, Neuropeptide Y/antagonists & inhibitors , Amides , Animals , Arginine/pharmacokinetics , Binding, Competitive , CHO Cells , Cell Line , Cricetinae , Cricetulus , Fluorescent Dyes , Fura-2 , Half-Life , Humans , Isotope Labeling , Molecular Probes , Neuropeptide Y/analogs & derivatives , Neuropeptide Y/pharmacology , Receptors, Neuropeptide/drug effects , Receptors, Neuropeptide Y/administration & dosage , Structure-Activity Relationship
11.
Endocrinology ; 156(12): 4639-48, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26418326

ABSTRACT

RF9, a reported antagonist of the mammalian gonadotropin-inhibitory hormone receptor, stimulates gonadotropin secretion in mammals. Recent studies have suggested that the stimulatory effect of RF9 on gonadotropin secretion relies on intact kisspeptin receptor (KISS1R) signaling, but the underlying mechanisms remain to be elucidated. Using Chinese Hamster Ovary cells stably transfected with KISS1R, we show that RF9 binds specifically to KISS1R, with a Kd of 1.6 × 10(-5)M, and stimulates an increase in intracellular calcium and inositol phosphate accumulation in a KISS1R-dependent manner, with EC50 values of 3.0 × 10(-6)M and 1.6 × 10(-7)M, respectively. RF9 also stimulated ERK phosphorylation, with a time course similar to that of kisspeptin-10. RFRP-3, the putative endogenous ligand for NPFFR1, did not stimulate inositol phosphate accumulation or pERK, nor did it alter responses to of kisspeptin-10 or RF9. In agreement with these in vitro data, we found that RF9 stimulated a robust LH increase in Npffr1(-/-) mice, similar to that in wild-type littermates, whereas the stimulatory effect of RF9 was markedly reduced in Kiss1r(-/-) and double Kiss1r(-/-)/Npfrr1(-/-) mice. The stimulatory effect of RF9 on LH secretion was restored by the selective rescue of Kiss1r expression in GnRH neurons, in Kiss1r(-/-T) mice. Taken together, our study demonstrates that RF9 acts primarily as a KISS1R agonist, but not as an allosteric modulator, to stimulate LH secretion. Our findings raise questions regarding the utility of RF9 for assessing NPFF1R function and de-emphasize a predominant role of this signaling system in central regulation of reproduction.


Subject(s)
Adamantane/analogs & derivatives , Dipeptides/pharmacology , Neuropeptides/pharmacology , Receptors, G-Protein-Coupled/drug effects , Receptors, Neuropeptide/drug effects , Adamantane/pharmacology , Animals , CHO Cells , Calcium/metabolism , Cricetulus , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , In Vitro Techniques , Inositol Phosphates/metabolism , Iodine Radioisotopes , Kisspeptins/metabolism , Luteinizing Hormone/drug effects , Luteinizing Hormone/metabolism , Mice , Mice, Knockout , Neurons/metabolism , Radioligand Assay , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/genetics , Receptors, Kisspeptin-1 , Receptors, Neuropeptide/genetics
12.
Oncotarget ; 6(30): 28573-87, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26378047

ABSTRACT

Preclinical evaluation of Retrocyclins (RC-100, RC-101) and Protegrin-1 (PG-1) antimicrobial peptides (AMPs) is important because of their therapeutic potential against bacterial, fungal and viral infections. Human mast cells (HMCs) play important roles in host defense and wound healing but the abilities of retrocyclins and protegrin-1 to harness these functions have not been investigated. Here, we report that chemically synthesized RC-100 and PG-1 caused calcium mobilization and degranulation in HMCs but these responses were not blocked by an inhibitor of formyl peptide receptor-like 1 (FPRL1), a known receptor for AMPs. However, RC-100 and PG-1 induced degranulation in rat basophilic leukemia (RBL-2H3) cells stably expressing Mas related G protein coupled receptor X2 (MrgX2). Chemical synthesis of these AMPs is prohibitively expensive and post-synthesis modifications (cyclization, disulfide bonds, folding) are inadequate for optimal antimicrobial activity. Indeed, we found that synthetic RC-100, which caused mast cell degranulation via MrgX2, did not display any antimicrobial activity. Green-fluorescent protein (GFP)-tagged RC-101 (analog of RC-100) and GFP-tagged PG-1 purified from transgenic plant chloroplasts killed bacteria and induced mast cell degranulation. Furthermore, GFP-PG1 bound specifically to RBL-2H3 cells expressing MrgX2. These findings suggest that retrocyclins and protegrins activate HMCs independently of FPRL1 but via MrgX2. Harnessing this novel feature of AMPs to activate mast cell's host defense/wound healing properties in addition to their antimicrobial activities expands their clinical potential. Low cost production of AMPs in plants should facilitate their advancement to the clinic overcoming major hurdles in current production systems.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Cell Degranulation/drug effects , Defensins/pharmacology , Immunologic Factors/pharmacology , Mast Cells/drug effects , Animals , Antimicrobial Cationic Peptides/biosynthesis , Antimicrobial Cationic Peptides/genetics , Bacteria/drug effects , Bacteria/growth & development , Calcium Signaling/drug effects , Cell Line, Tumor , Defensins/biosynthesis , Defensins/genetics , Dose-Response Relationship, Drug , Humans , Immunologic Factors/biosynthesis , Immunologic Factors/genetics , Mast Cells/immunology , Mast Cells/metabolism , Microbial Viability/drug effects , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Plants, Genetically Modified/genetics , Plants, Genetically Modified/metabolism , Rats , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/drug effects , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism , Recombinant Fusion Proteins/pharmacology , Signal Transduction/drug effects , Time Factors , Transfection
13.
Neuro Endocrinol Lett ; 36(8): 767-70, 2015.
Article in English | MEDLINE | ID: mdl-26921577

ABSTRACT

OBJECTIVE: The neonatal and/or prepubertal androgen milieu affects sexual maturation. In rodents, neonatal chronic testosterone treatment, which is used as a model of polycystic ovary syndrome (PCOS), results in the onset of vaginal opening occurring earlier in the pubertal period. DESIGN: In the present study, the changes in hypothalamic Kiss1 (a gonadotropin-releasing hormone (GnRH)-stimulating factor) and RF-amide related peptide (RFRP; a GnRH inhibitory factor) mRNA expression induced by testosterone treatment were examined in order to clarify whether these factors are involved in the testosterone-induced acceleration of sexual maturation. RESULTS: The onset of vaginal opening occurred earlier and uterine weight was increased in female rats subjected to chronic (from postnatal day 23 to day 31) testosterone treatment. Contrary to our expectations, the rats' hypothalamic Kiss1 and Kiss1 receptor mRNA levels were not changed, and their serum luteinizing hormone (LH) levels were decreased. Although hypothalamic RFRP mRNA expression was decreased in the testosterone-treated rats, this change was not reflected in their serum LH levels. CONCLUSIONS: These results indicate that the advancement of sexual maturation observed in chronic testosterone-treated rats might be caused by a peripheral, rather than a central, mechanism.


Subject(s)
Androgens/pharmacology , Hypothalamus/drug effects , Kisspeptins/drug effects , Neuropeptides/drug effects , RNA, Messenger/drug effects , Sexual Maturation/drug effects , Testosterone/pharmacology , Vagina/drug effects , Animals , Female , Gonadotropin-Releasing Hormone/drug effects , Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/metabolism , Kisspeptins/genetics , Kisspeptins/metabolism , Neuropeptides/genetics , Neuropeptides/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Kisspeptin-1 , Receptors, Neuropeptide/drug effects , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism
14.
Brain Res ; 1508: 73-82, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23500635

ABSTRACT

Orexin and its receptors (OxR1 and OxR2) play a significant role in arousal and sleep regulation. Using developing piglets, we aimed to determine the effects of nicotine and Intermittent Hypercapnic Hypoxia (IHH), alone or in combination, on orexin receptor expression in the hypothalamus. Four piglet groups were studied: control (n=14), nicotine (n=14), IHH (n=10) and nic+IHH (n=14). Applying immunohistochemistry for OxR1 and OxR2 expression, eight nuclei/areas of the hypothalamus: dorsal medial nucleus (DMN), arcuate nucleus (ARC), perifornical area (PFA), paraventricular nucleus (PVN), lateral hypothalamic area (LHA), ventral medial nucleus (VMN), supraoptic nucleus, retrochiasmatic part (SONr) and tuberal mammillary nucleus (TMN), were studied. Compared to controls, OxR1 and OxR2 were increased due to exposures, however this was region dependent. Nicotine increased OxR1 in the DMN (P<0.001) and SONr (P=0.036), and OxR2 in the DMN (P<0.001), VMN (P=0.014) and the TMN (P=0.026). IHH increased OxR1 in the DMN, PVN, VMN and SONr (P<0.01 for all), and OxR2 in DMN (P<0.001), PFA (P=0.001), PVN (P=0.004), VMN (P=0.041) and the TMN (P<0.001). The nic+IHH exposure increased OxR1 expression in all nuclei (TMN excluded) however, the changes were not significantly different from IHH alone. For OxR2, the increased expression after nic+IHH was significant compared to IHH in the DMN, ARC and SONr. These results show that nicotine increases orexin receptor expression in a region dependent manner. IHH induced increases were specific to arousal and stress related regions and nic+IHH results suggest that for OxR1, nicotine has no additive effect whereas for OxR2 it does, and is region dependent.


Subject(s)
Hypercapnia/metabolism , Hypothalamus/growth & development , Hypothalamus/metabolism , Hypoxia/metabolism , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/metabolism , Animals , Arousal/physiology , Female , Humans , Hypothalamus/drug effects , Immunohistochemistry , Infant, Newborn , Male , Orexin Receptors , Prone Position , Receptors, G-Protein-Coupled/drug effects , Receptors, Neuropeptide/drug effects , Sex Characteristics , Sleep/physiology , Sleep Apnea, Obstructive/pathology , Sudden Infant Death , Swine
15.
Learn Mem ; 20(3): 139-46, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23418393

ABSTRACT

The allatostatin receptor (AlstR)/ligand inactivation system enables potent regulation of neuronal circuit activity. To examine how different cell types participate in memory formation, we have used this system through Cre-directed, cell-type specific expression in mouse hippocampal CA1 in vivo and examined functional effects of inactivation of excitatory vs. inhibitory neurons on memory formation. We chose to use a hippocampus-dependent behavioral task involving location-dependent object recognition (LOR). The double transgenic mice, with the AlstRs selectively expressed in excitatory pyramidal neurons or inhibitory interneurons, were cannulated, targeting dorsal hippocampus to allow the infusion of the receptor ligand (the allatostatin [AL] peptide) in a time dependent manner. Compared to control animals, AL-infused animals showed no long-term memory for object location. While inactivation of excitatory or inhibitory neurons produced opposite effects on hippocampal circuit activity in vitro, the effects in vivo were similar. Both types of inactivation experiments resulted in mice exhibiting no long-term memory for object location. Together, these results demonstrate that the Cre-directed, AlstR-based system is a powerful tool for cell-type specific manipulations in a behaving animal and suggest that activity of either excitatory neurons or inhibitory interneurons is essential for proper long-term object location memory formation.


Subject(s)
CA1 Region, Hippocampal/drug effects , Receptors, Neuropeptide/drug effects , Recognition, Psychology/drug effects , Animals , CA1 Region, Hippocampal/cytology , Catheterization , Interneurons/drug effects , Ligands , Memory/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Net/cytology , Nerve Net/drug effects , Orientation/drug effects , Pyramidal Cells/drug effects , Receptors, Neuropeptide/genetics
16.
Mol Cell Endocrinol ; 371(1-2): 189-94, 2013 May 22.
Article in English | MEDLINE | ID: mdl-23267837

ABSTRACT

Pheochromocytoma is a rare but potentially lethal chromaffin cell tumor with currently no effective treatment. Peptide hormone receptors are frequently overexpressed on endocrine tumor cells and can be specifically targeted by various anti-tumor peptide analogs. The present study carried out on mouse pheochromocytoma cells (MPCs) and a more aggressive mouse tumor tissue-derived (MTT) cell line revealed that these cells are characterized by pronounced expression of the somatostatin receptor 2 (sst2), growth hormone-releasing hormone (GHRH) receptor and the luteinizing hormone-releasing hormone (LHRH) receptor. We further demonstrated significant anti-tumor effects mediated by cytotoxic somatostatin analogs, AN-162 and AN-238, by LHRH antagonist, Cetrorelix, by the cytotoxic LHRH analog, AN-152, and by recently developed GHRH antagonist, MIA-602, on MPC and for AN-152 and MIA-602 on MTT cells. Studies of novel anti-tumor compounds on these mouse cell lines serve as an important basis for mouse models of metastatic pheochromocytoma, which we are currently establishing.


Subject(s)
Adrenal Gland Neoplasms/drug therapy , Pheochromocytoma/drug therapy , Receptors, Neuropeptide/drug effects , 2-Hydroxyphenethylamine/analogs & derivatives , 2-Hydroxyphenethylamine/pharmacology , Aniline Compounds/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Doxorubicin/analogs & derivatives , Doxorubicin/pharmacology , Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Gonadotropin-Releasing Hormone/pharmacology , Growth Hormone-Releasing Hormone/antagonists & inhibitors , Mice , Pyrroles/pharmacology , Receptors, LHRH/biosynthesis , Receptors, LHRH/drug effects , Receptors, LHRH/metabolism , Receptors, Neuropeptide/biosynthesis , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/biosynthesis , Receptors, Pituitary Hormone-Regulating Hormone/drug effects , Receptors, Pituitary Hormone-Regulating Hormone/metabolism , Receptors, Somatostatin/biosynthesis , Receptors, Somatostatin/drug effects , Receptors, Somatostatin/metabolism , Sermorelin/analogs & derivatives , Sermorelin/pharmacology , Somatostatin/analogs & derivatives
17.
Psychopharmacology (Berl) ; 226(4): 687-98, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22411428

ABSTRACT

RATIONALE: Glutamate and orexin/hypocretin systems are involved in Pavlovian cue-triggered drug seeking. OBJECTIVES: Here, we asked whether orexin and glutamate interact within ventral tegmental area (VTA) to promote reinstatement of extinguished cocaine seeking in a rat self-administration paradigm. METHODS/RESULTS: We first found that bilateral VTA microinjections of the orexin 1 receptor (OX1R) antagonist SB-334867 (SB) or a cocktail of the AMPA and NMDA glutamate receptor antagonists CNQX/AP-5 reduced reinstatement of cocaine seeking elicited by cues. In contrast, neither of these microinjections nor systemic SB reduced cocaine-primed reinstatement. Additionally, unilateral VTA OX1R blockade combined with contralateral VTA glutamate blockade attenuated cue-induced reinstatement, indicating that VTA orexin and glutamate are simultaneously necessary for cue-induced reinstatement. We further probed the receptor specificity of glutamate actions in VTA, finding that CNQX, but not AP-5, dose-dependently attenuated cue-induced reinstatement, indicating that AMPA but not NMDA receptor transmission is required for this type of cocaine seeking. Given the necessary roles of both OX1 and AMPA receptors in VTA for cue-induced cocaine seeking, we hypothesized that these signaling pathways interact during this behavior. We found that PEPA, a positive allosteric modulator of AMPA receptors, completely reversed the SB-induced attenuation of reinstatement behavior. Intra-VTA PEPA alone did not alter cue-induced reinstatement, indicating that potentiating AMPA activity with this drug specifically compensates for OX1R blockade, rather than simply inducing or enhancing reinstatement itself. CONCLUSIONS: These findings show that cue-induced, but not cocaine-primed, reinstatement of cocaine seeking is dependent upon orexin and AMPA receptor interactions in VTA.


Subject(s)
Cocaine/administration & dosage , Glutamic Acid/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Neuropeptides/metabolism , Ventral Tegmental Area/metabolism , 2-Amino-5-phosphonovalerate/administration & dosage , 2-Amino-5-phosphonovalerate/pharmacology , 6-Cyano-7-nitroquinoxaline-2,3-dione/administration & dosage , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Benzoxazoles/pharmacology , Cues , Dose-Response Relationship, Drug , Drug-Seeking Behavior , Male , Naphthyridines , Orexin Receptors , Orexins , Rats , Rats, Sprague-Dawley , Receptors, AMPA/antagonists & inhibitors , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, Neuropeptide/drug effects , Receptors, Neuropeptide/metabolism , Self Administration , Signal Transduction , Urea/analogs & derivatives , Urea/pharmacology
18.
Psychopharmacology (Berl) ; 226(2): 347-55, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23149909

ABSTRACT

RATIONALE: Previous studies have shown that activation of brain neuropeptide S receptor (NPSR) facilitates reinstatement of cocaine seeking elicited by environmental cues predictive of drug availability. This finding suggests the possibility that blockade of NPSR receptors may be of therapeutic benefit in cocaine addiction. To evaluate this hypothesis, we investigated the effect of two newly synthetized NPSR antagonists, namely the quinolinone-amide derivative NPSR-QA1 and the NPS peptidic analogue [D-Cys(tBu)5]NPS on cocaine self-administration and on discriminative cue-induced relapse to cocaine seeking in the rat. METHODS: Separate groups of rats self-administered food and cocaine 0.25 mg/kg/inf in FR1 and FR5 (fixed ratio reinforcement schedules) for 30-min and 2-h sessions per day. After food and cocaine intake reached baseline levels, the effect of NPSR-QA1 was tested on cocaine and food self-administration. The NPSR-QA1 was injected intraperitoneally and its effect on discriminative cue-induced reinstatement was evaluated, while [D-Cys(tBut)5]NPS was injected intracranially, intra-lateral hypothalamus, intra-perifornical area of the hypothalamus, and intra-central amygdala. The effect of the NPSR-QA1 on extinction of cocaine seeking was also assessed. RESULTS: Intraperitoneal administration of NPSR-QA1 (15-30 mg/kg) did not affect cocaine self-administration. Conversely, NPSR-QA1 (15-30 mg/kg) decreased discriminative cue-induced cocaine relapse. At the lowest dose, this effect was specific, while at the highest dose, NPSR-QA1 also reduced food self-administration. The efficacy of NPSR antagonism on cocaine seeking was confirmed with [D-Cys(tBu)5]NPS (10-30 nmol/rat) as it markedly inhibited relapse behavior following site-specific injection into the lateral hypothalamus and the perifornical area of the hypothalamus but not into the central amygdala. CONCLUSIONS: The identification of the NPS/NPSR system as an important new element involved in the physiopathology of cocaine addiction and the discovery of the anti-addictive properties of NPSR antagonists opens the possibility of exploring a new mechanism for cocaine addiction treatment.


Subject(s)
Cocaine/pharmacology , Cues , Hypothalamus/metabolism , Neuropeptides/antagonists & inhibitors , Receptors, Neuropeptide/drug effects , Amides/pharmacology , Animals , Behavior, Addictive/drug therapy , Dose-Response Relationship, Drug , Drug-Seeking Behavior/drug effects , Male , Neuropeptides/pharmacology , Quinolones/pharmacology , Rats , Rats, Wistar
19.
Neuroscience ; 226: 81-8, 2012 Dec 13.
Article in English | MEDLINE | ID: mdl-22986171

ABSTRACT

UNLABELLED: Very recently, we have reported that the modulatory effect of PVN on gastric acid secretion may be mediated through the orexin fibers and/or orexin-responsive neurons. In this study, we address the hypothesis which demonstrates the existence of a putative orexin A - neuropeptide Y Y1/Y5 receptors interaction to increase gastric acid secretion in pyloric-ligated conscious rats. Male Wistar rats were implanted with guide canula directed to the PVN and lateral ventricle. Intracerebroventricular (ICV) microinjections of GR-231118 (Y1 receptor antagonist) and CGP-71683 (Y5 receptor antagonist) on gastric acid secretion were considered. The effect of pretreatment with Y1 receptor antagonist, GR-231118, and Y5 receptor antagonist, CGP-71683, on PVN orexin A-induced acid secretion was assessed. Gastric acid secretion was measured using the pylorus-ligation method, and the amount of gastric acid was determined by titration with 0.01N NaOH to a pH of 7.0. KEY RESULTS: ICV microinjections of GR-231118 and CGP-71683 decreased acid secretion by 25±0.05% and 67±0.02%, respectively. ICV microinjections of GR-231118 and CGP-71683 inhibited effects of PVN-injected orexin-A on acid secretion. We suggest that Y1 and Y5 receptors stimulate gastric acid secretion and the stimulatory effect of PVN orexin receptors on gastric acid secretion may be mediated via interactions, at least in part, through activation of Y1 and Y5 receptors. These neural pathways may play key roles in the orexinergic action of orexins in the cephalic phase of gastric acid secretion.


Subject(s)
Gastric Acid/metabolism , Intracellular Signaling Peptides and Proteins/pharmacology , Neuropeptide Y/physiology , Neuropeptides/pharmacology , Paraventricular Hypothalamic Nucleus/drug effects , Animals , Gastric Juice/metabolism , Intracellular Signaling Peptides and Proteins/administration & dosage , Male , Microinjections , Naphthalenes/administration & dosage , Naphthalenes/pharmacology , Neuropeptide Y/antagonists & inhibitors , Neuropeptides/administration & dosage , Orexins , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/pharmacology , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/drug effects , Receptors, Neuropeptide/antagonists & inhibitors , Receptors, Neuropeptide/drug effects , Receptors, Neuropeptide Y/antagonists & inhibitors , Receptors, Neuropeptide Y/drug effects
20.
Curr Pharm Des ; 18(35): 5709-27, 2012.
Article in English | MEDLINE | ID: mdl-22632474

ABSTRACT

In addition to the classical neurotransmitters, neuropeptides represent an important class of modulators for affective behaviors and associated disorders, such as anxiety disorders. Many neuropeptides are abundantly expressed in brain regions involved in emotional processing and anxiety behaviors. Moreover, risk factors for anxiety disorders such as stress modulate the expression of various neuropeptides in the brain. Due to the high prevalence of anxiety disorders and yet limited treatment options, there is a clear need for more effective therapeutics. In this regard, the various neuropeptides represent exciting candidates for new therapeutic designs. In this review, I will provide an up-to-date summary on the evidences for the involvement of seven neuropeptides in anxiety: corticotropin-releasing factor, urocortins, vasopressin, oxytocin, substance P, neuropeptide Y and galanin. This review will cover the behavioral effects of these neuropeptides in animal models of anxiety by both genetic and pharmacological manipulations. Human studies indicating a role for these neuropeptides in anxiety disorders will also be discussed.


Subject(s)
Anxiety Disorders/physiopathology , Drug Design , Neuropeptides/metabolism , Animals , Anxiety Disorders/drug therapy , Brain/physiopathology , Humans , Molecular Targeted Therapy , Receptors, Neuropeptide/drug effects , Receptors, Neuropeptide/metabolism , Risk Factors , Stress, Physiological , Stress, Psychological/complications
SELECTION OF CITATIONS
SEARCH DETAIL
...