Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 213
Filter
1.
Pharmacol Res ; 203: 107173, 2024 May.
Article in English | MEDLINE | ID: mdl-38580186

ABSTRACT

Our recent multi-omics studies have revealed rich sources of novel bioactive proteins and polypeptides from marine organisms including cnidarians. In the present study, we initially conducted a transcriptomic analysis to review the composition profile of polypeptides from Zoanthus sociatus. Then, a newly discovered NPY-like polypeptide-ZoaNPY was selected for further in silico structural, binding and virtually pharmacological studies. To evaluate the pro-angiogenic effects of ZoaNPY, we employed an in vitro HUVECs model and an in vivo zebrafish model. Our results indicate that ZoaNPY, at 1-100 pmol, enhances cell survival, migration and tube formation in the endothelial cells. Besides, treatment with ZoaNPY could restore a chemically-induced vascular insufficiency in zebrafish embryos. Western blot results demonstrated the application of ZoaNPY could increase the phosphorylation of proteins related to angiogenesis signaling including PKC, PLC, FAK, Src, Akt, mTOR, MEK, and ERK1/2. Furthermore, through molecular docking and surface plasmon resonance (SPR) verification, ZoaNPY was shown to directly and physically interact with NPY Y2 receptor. In view of this, all evidence showed that the pro-angiogenic effects of ZoaNPY involve the activation of NPY Y2 receptor, thereby activating the Akt/mTOR, PLC/PKC, ERK/MEK and Src- FAK-dependent signaling pathways. Furthermore, in an excision wound model, the treatment with ZoaNPY was shown to accelerate the wound healing process in mice. Our findings provide new insights into the discovery and development of novel pro-angiogenic drugs derived from NPY-like polypeptides in the future.


Subject(s)
Cnidaria , Peptides , Receptors, Neuropeptide Y , Animals , Humans , Mice , Cell Movement/drug effects , Focal Adhesion Kinase 1/drug effects , Focal Adhesion Kinase 1/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Ligands , Molecular Docking Simulation , Neovascularization, Physiologic/drug effects , Neuropeptide Y/metabolism , Neuropeptide Y/pharmacology , Peptides/pharmacology , Protein Kinase C/drug effects , Protein Kinase C/metabolism , Receptors, Neuropeptide Y/drug effects , Receptors, Neuropeptide Y/metabolism , Signal Transduction/drug effects , src-Family Kinases/drug effects , src-Family Kinases/metabolism , Zebrafish , Cnidaria/chemistry , Phosphoinositide Phospholipase C/drug effects , Phosphoinositide Phospholipase C/metabolism
2.
Proc Natl Acad Sci U S A ; 117(27): 15862-15873, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32561647

ABSTRACT

Albuminuria is an independent risk factor for the progression to end-stage kidney failure, cardiovascular morbidity, and premature death. As such, discovering signaling pathways that modulate albuminuria is desirable. Here, we studied the transcriptomes of podocytes, key cells in the prevention of albuminuria, under diabetic conditions. We found that Neuropeptide Y (NPY) was significantly down-regulated in insulin-resistant vs. insulin-sensitive mouse podocytes and in human glomeruli of patients with early and late-stage diabetic nephropathy, as well as other nondiabetic glomerular diseases. This contrasts with the increased plasma and urinary levels of NPY that are observed in such conditions. Studying NPY-knockout mice, we found that NPY deficiency in vivo surprisingly reduced the level of albuminuria and podocyte injury in models of both diabetic and nondiabetic kidney disease. In vitro, podocyte NPY signaling occurred via the NPY2 receptor (NPY2R), stimulating PI3K, MAPK, and NFAT activation. Additional unbiased proteomic analysis revealed that glomerular NPY-NPY2R signaling predicted nephrotoxicity, modulated RNA processing, and inhibited cell migration. Furthermore, pharmacologically inhibiting the NPY2R in vivo significantly reduced albuminuria in adriamycin-treated glomerulosclerotic mice. Our findings suggest a pathogenic role of excessive NPY-NPY2R signaling in the glomerulus and that inhibiting NPY-NPY2R signaling in albuminuric kidney disease has therapeutic potential.


Subject(s)
Albuminuria/metabolism , Kidney Diseases/metabolism , Neuropeptide Y/metabolism , Receptors, Neuropeptide Y/metabolism , Signal Transduction/physiology , Animals , Arginine/analogs & derivatives , Arginine/pharmacology , Benzazepines/pharmacology , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies , Disease Models, Animal , Down-Regulation , Doxorubicin/pharmacology , Humans , Insulin/metabolism , Kidney Diseases/pathology , Kidney Glomerulus/drug effects , Kidney Glomerulus/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Neuropeptide Y/pharmacology , Neuropeptide Y/urine , Podocytes/metabolism , Proteomics , Receptors, Neuropeptide Y/drug effects , Signal Transduction/drug effects
3.
Am J Physiol Regul Integr Comp Physiol ; 316(4): R406-R416, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30726118

ABSTRACT

Neuropeptide Y (NPY), peptide YY (PYY), and their cognate receptors (YR) are expressed by subpopulations of central and peripheral nervous system neurons. Intracerebroventricular injections of NPY or PYY increase food intake, and intrahypothalamic NPY1 or NPY5 receptor agonist injections also increase food intake. In contrast, injection of PYY in the periphery reduces food intake, apparently by activating peripheral Y2R. The dorsal vagal complex (DVC) of the hindbrain is the site where vagal afferents relay gut satiation signals to the brain. While contributions of the DVC are increasingly investigated, a role for DVC YR in control of food intake has not been examined systematically. We used in situ hybridization to confirm expression of Y1R and Y2R, but not Y5R, in the DVC and vagal afferent neurons. We found that nanoinjections of a Y2R agonist, PYY-(3-36), into the DVC significantly increased food intake over a 4-h period in satiated male rats. PYY-(3-36)-evoked food intake was prevented by injection of a selective Y2R antagonist. Injection of a Y1R/Y5R-preferring agonist into the DVC failed to increase food intake at doses reported to increase food intake following hypothalamic injection. Finally, injection of PYY-(3-36) into the DVC prevented reduction of 30-min food intake following intraperitoneal injection of cholecystokinin (CCK). Our results indicate that activation of DVC Y2R, unlike hypothalamic or peripheral Y2R, increases food intake. Furthermore, in the context of available electrophysiological observations, our results are consistent with the hypothesis that DVC Y2R control food intake by dampening vagally mediated satiation signals in the DVC.


Subject(s)
Cholecystokinin/pharmacology , Eating/drug effects , Receptors, Neuropeptide Y/agonists , Satiation/drug effects , Vagus Nerve/drug effects , Animals , Injections , Male , Peptide YY/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Neuropeptide Y/antagonists & inhibitors , Receptors, Neuropeptide Y/drug effects
4.
Addict Biol ; 21(4): 755-65, 2016 07.
Article in English | MEDLINE | ID: mdl-25904345

ABSTRACT

Neuropeptide Y (NPY), which is widely expressed in the central nervous system is involved in several neuropathologies including addiction. Here we comprehensively and systematically review alterations on the central NPY system induced by several drugs. We report on the effects of psychostimulants [cocaine, amphetamine, methamphetamine, 3,4-methylenedioxymethamphetamine (MDMA) and nicotine], ethanol, and opioids on NPY protein levels and expression of different NPY receptors. Overall, expression and function of NPY and its receptors are changed under conditions of drug exposure, thus affecting several physiologic behaviors, such as feeding, stress and anxiety. Drugs of abuse differentially affect the components of the NPY system. For example methamphetamine and nicotine lead to a consistent increase in NPY mRNA and protein levels in different brain sites whereas ethanol and opioids decrease NPY mRNA and protein expression. Drug-induced alterations on the different NPY receptors show more complex regulation pattern. Manipulation of the NPY system can have opposing effects on reinforcing and addictive properties of drugs of abuse. NPY can produce pro-addictive effects (nicotine and heroin), but can also exert inhibitory effects on addictive behavior (AMPH, ethanol). Furthermore, NPY can act as a neuroprotective agent in chronically methamphetamine and MDMA-treated rodents. In conclusion, manipulation of the NPY system seems to be a potential target to counteract neural alterations, addiction-related behaviors and cognitive deficits induced by these drugs.


Subject(s)
Amphetamines/pharmacology , Cocaine/pharmacology , Ethanol/pharmacology , Neuropeptide Y/drug effects , Nicotine/pharmacology , Opiate Alkaloids/pharmacology , Animals , Brain/drug effects , Disease Models, Animal , Mice , Rats , Receptors, Neuropeptide Y/drug effects
5.
Am J Physiol Endocrinol Metab ; 309(8): E759-66, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26330345

ABSTRACT

Although central PYY delivery potently increases food intake, the sites of action and mechanisms mediating these hyperphagic effects are not fully understood. The present studies investigate the contribution of lateral parabrachial nucleus (lPBN) PYY-Y receptor signaling to food intake control, as lPBN neurons express Y receptors and receive PYY fibers and are known to integrate circulating and visceral sensory signals to regulate energy balance. Immunohistochemical results identified a subpopulation of gigantocellular reticular nucleus PYY-producing neurons that project monosynaptically to the lPBN, providing an endogenous source of PYY to the lPBN. lPBN microinjection of PYY-(1-36) or PYY-(3-36) markedly increased food intake by increasing meal size. To determine which receptors mediate these behavioral results, we first performed quantitative real-time PCR to examine the relative levels of Y receptor expression in lPBN tissue. Gene expression analyses revealed that, while Y1, Y2, and Y5 receptors are each expressed in lPBN tissue, Y1 receptor mRNA is expressed at fivefold higher levels than the others. Furthermore, behavioral/pharmacological results demonstrated that the hyperphagic effects of PYY-(3-36) were eliminated by lPBN pretreatment with a selective Y1 receptor antagonist. Together, these results highlight the lPBN as a novel site of action for the intake-stimulatory effects of central PYY-Y1 receptor signaling.


Subject(s)
Feeding Behavior/physiology , Parabrachial Nucleus/metabolism , Peptide Fragments/metabolism , Peptide YY/metabolism , RNA, Messenger/metabolism , Receptors, Neuropeptide Y/genetics , Animals , Feeding Behavior/drug effects , Male , Microinjections , Parabrachial Nucleus/drug effects , Parabrachial Nucleus/physiology , Peptide Fragments/pharmacology , Peptide YY/pharmacology , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Receptors, Neuropeptide Y/drug effects , Receptors, Neuropeptide Y/metabolism
6.
J Physiol ; 592(7): 1655-75, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24535439

ABSTRACT

Neuropeptide Y (NPY), a brain neuromodulator that has been strongly implicated in the regulation of energy balance, also acts centrally to inhibit sympathetic nerve activity (SNA); however, the site and mechanism of action are unknown. In chloralose-anaesthetized female rats, nanoinjection of NPY into the paraventricular nucleus of the hypothalamus (PVN) dose-dependently suppressed lumbar SNA (LSNA) and its baroreflex regulation, and these effects were blocked by prior inhibition of NPY Y1 or Y5 receptors. Moreover, PVN injection of Y1 and Y5 receptor antagonists in otherwise untreated rats increased basal and baroreflex control of LSNA, indicating that endogenous NPY tonically inhibits PVN presympathetic neurons. The sympathoexcitation following blockade of PVN NPY inhibition was eliminated by prior PVN nanoinjection of the melanocortin 3/4 receptor inhibitor SHU9119. Moreover, presympathetic neurons, identified immunohistochemically using cholera toxin b neuronal tract tracing from the rostral ventrolateral medulla (RVLM), express NPY Y1 receptor immunoreactivity, and patch-clamp recordings revealed that both NPY and α-melanocyte-stimulating hormone (α-MSH) inhibit and stimulate, respectively, PVN-RVLM neurons. Collectively, these data suggest that PVN NPY inputs converge with α-MSH to influence presympathetic neurons. Together these results identify endogenous NPY as a novel and potent inhibitory neuromodulator within the PVN that may contribute to changes in SNA that occur in states associated with altered energy balance, such as obesity and pregnancy.


Subject(s)
Baroreflex/drug effects , Neural Inhibition/drug effects , Neuropeptide Y/pharmacology , Paraventricular Hypothalamic Nucleus/drug effects , Sympathetic Nervous System/drug effects , Animals , Dose-Response Relationship, Drug , Evoked Potentials , Female , Injections , Male , Neuropeptide Y/administration & dosage , Paraventricular Hypothalamic Nucleus/physiology , Rats, Sprague-Dawley , Rats, Wistar , Receptors, Neuropeptide Y/drug effects , Receptors, Neuropeptide Y/metabolism , Sympathetic Nervous System/physiology , Time Factors , alpha-MSH/pharmacology
7.
J Neurochem ; 120(1): 93-105, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22007767

ABSTRACT

Increasing evidences suggest that neuropeptide Y (NPY) may act as a key modulator of the cross-talk between the brain and the immune system in health and disease. In the present study, we dissected the possible inhibitory role of NPY upon inflammation-associated microglial cell motility. NPY, through activation of Y(1) receptors, was found to inhibit lipopolysaccharide (LPS)-induced microglia (N9 cell line) motility. Moreover, stimulation of microglia with LPS was inhibited by IL-1 receptor antagonist (IL-1ra), suggesting the involvement of endogenous interleukin-1 beta (IL-1ß) in this process. Direct stimulation with IL-1ß promoted downstream p38 mitogen-activated protein kinase mobilization and increased microglia motility. Moreover, consistently, p38 mitogen-activated protein kinase inhibition decreased the extent of actin filament reorganization occurring during plasma membrane ruffling and p38 phosphorylation was inhibited by NPY, involving Y(1) receptors. Significantly, the key inhibitory role of NPY on LPS-induced motility of CD11b-positive cells was further confirmed in mouse brain cortex explants. In summary, we revealed a novel functional role for NPY in the regulation of microglial function that may have important implications in the modulation of CNS injuries/diseases where microglia migration/motility might play a role.


Subject(s)
Cell Movement/drug effects , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/pharmacology , Microglia/drug effects , Neuropeptide Y/pharmacology , Animals , Blotting, Western , CD11b Antigen/metabolism , Cell Line , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cytoskeleton/drug effects , Cytoskeleton/ultrastructure , Data Interpretation, Statistical , Immunohistochemistry , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Receptors, Neuropeptide Y/drug effects , p38 Mitogen-Activated Protein Kinases/physiology
8.
J Neurochem ; 122(2): 415-26, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22537092

ABSTRACT

The effects of intracerebroventricular administration of neuropeptide Y (NPY), which is believed to play an important role in neuroprotection against excitotoxicity and in the modulation of adult neurogenesis, were evaluated in an animal model of hippocampal neurodegeneration and temporal lobe epilepsy represented by trimethyltin (TMT) intoxication. A single TMT injection (8 mg/kg) causes, in the rat brain, massive neuronal death, selectively involving pyramidal neurons, accompanied by glial activation and enhanced hippocampal neurogenesis. Our data indicate that intracerebroventricular administration of exogenous NPY (at the dose of 2 µg/2 µL, 4 days after TMT-administration), in adult rats, exerts a protective role in regard to TMT-induced hippocampal damage and a proliferative effect on the hippocampal neurogenic niche through the up-regulation of Bcl-2, Bcl2l1, Bdnf, Sox-2, NeuroD1, Noggin and Doublecortin genes, contributing to delineate more clearly the role of NPY in in vivo neurodegenerative processes.


Subject(s)
Epilepsy, Temporal Lobe/drug therapy , Epilepsy, Temporal Lobe/pathology , Hippocampus/drug effects , Nerve Degeneration/prevention & control , Neurogenesis/drug effects , Neuropeptide Y/pharmacology , Neuroprotective Agents , Trimethyltin Compounds , Animals , Antimetabolites , Apoptosis Regulatory Proteins/biosynthesis , Brain-Derived Neurotrophic Factor/biosynthesis , Bromodeoxyuridine , Doublecortin Protein , Epilepsy, Temporal Lobe/chemically induced , Female , Gene Expression/drug effects , Hippocampus/pathology , Immunohistochemistry , Injections, Intraventricular , Nerve Degeneration/etiology , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neurogenesis/genetics , Neuropeptide Y/administration & dosage , RNA/biosynthesis , RNA/isolation & purification , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Receptors, Neuropeptide Y/drug effects
9.
Pharm Res ; 29(6): 1698-711, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22399387

ABSTRACT

PURPOSE: Pancreatic polypeptide (PP) has important glucoregulatory functions and thereby holds significance in the treatment of diabetes and obesity. However, short plasma half-life and aggregation propensity of PP in aqueous solution, limits its therapeutic application. To address these issues, we prepared and characterized a formulation of PP in sterically stabilized micelles (SSM) that protects and stabilizes PP in its active conformation. METHODS: PP-SSM was prepared by incubating PP with SSM dispersion in buffer. Peptide-micelle association and freeze-drying efficacy of the formulation was characterized in phosphate buffers with or without sodium chloride using dynamic light scattering, fluorescence spectroscopy and circular dichroism. The degradation kinetics of PP-SSM in presence of proteolytic enzyme was determined using HPLC and bioactivity of the formulation was evaluated by in vitro cAMP inhibition study. RESULTS: PP self-associated with SSM and this interaction was influenced by presence/absence of sodium chloride in the buffer. The formulation was effectively lyophilized, demonstrating feasibility for its long-term storage. The stability of peptide against proteolytic degradation was significantly improved and PP in SSM retained its bioactivity in vitro. CONCLUSIONS: Self-association of PP with phospholipid micelles addressed the delivery issues of the peptide. This nanomedicine should be further developed for the treatment of diabetes.


Subject(s)
Drug Carriers , Hypoglycemic Agents/chemistry , Micelles , Pancreatic Polypeptide/chemistry , Phosphatidylethanolamines/chemistry , Polyethylene Glycols/chemistry , Protein Precursors/chemistry , Cell Line, Tumor , Chemistry, Pharmaceutical , Chromatography, High Pressure Liquid , Circular Dichroism , Colforsin/pharmacology , Cyclic AMP/metabolism , Drug Compounding , Drug Stability , Freeze Drying , Humans , Hydrogen-Ion Concentration , Light , Nanoparticles , Nanotechnology , Neuroblastoma/metabolism , Particle Size , Protein Denaturation , Receptors, Neuropeptide Y/drug effects , Receptors, Neuropeptide Y/metabolism , Scattering, Radiation , Sodium Chloride/chemistry , Spectrometry, Fluorescence , Technology, Pharmaceutical/methods , Time Factors
10.
Hippocampus ; 21(2): 198-211, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20043282

ABSTRACT

Cholecystokinin (CCK)-expressing basket cells encompass a subclass of inhibitory GABAergic interneurons that regulate memory-forming oscillatory network activity of the hippocampal formation in accordance to the emotional and motivational state of the animal, conveyed onto these cells by respective extrahippocampal afferents. Various excitatory and inhibitory afferent and efferent synapses of the hippocampal CCK basket cells express serotoninergic, cholinergic, cannabinoid, and benzodiazepine sensitive receptors, all contributing to their functional plasticity. We explored whether CCK basket cells are modulated by neuropeptide Y (NPY), one of the major local neuropeptides that strongly inhibits hippocampal excitability and has significant effect on its memory function. Here, using GAD65-GFP transgenic mice for prospective identification of CCK basket cells and whole-cell patch-clamp recordings, we show for the first time that excitatory and inhibitory inputs onto CCK basket cells in the dentate gyrus of the hippocampus are modulated by NPY through activation of NPY Y2 receptors. The frequency of spontaneous and miniature EPSCs, as well as the amplitudes of stimulation-evoked EPSCs were decreased. Similarly, the frequency of both spontaneous and miniature IPSCs, and the amplitudes of stimulation-evoked IPSCs were decreased after NPY application. Most of the effects of NPY could be attributed to a presynaptic site of action. Our data provide the first evidence that the excitatory and inhibitory inputs onto the CCK basket cells could be modulated by local levels of NPY, and may change the way these cells process extrahippocampal afferent information, influencing hippocampal function and its network excitability during normal and pathological oscillatory activities.


Subject(s)
Hippocampus/physiology , Neuropeptide Y/physiology , Animals , Cholecystokinin/physiology , Dentate Gyrus/cytology , Dentate Gyrus/drug effects , Dentate Gyrus/physiology , Electrophysiological Phenomena , Excitatory Postsynaptic Potentials/physiology , Glutamate Decarboxylase/genetics , Hippocampus/cytology , Hippocampus/drug effects , In Vitro Techniques , Inhibitory Postsynaptic Potentials/physiology , Interneurons/drug effects , Interneurons/physiology , Mice , Mice, Transgenic , Miniature Postsynaptic Potentials/physiology , Neurons, Afferent/physiology , Neuropeptide Y/pharmacology , Patch-Clamp Techniques , Receptors, Neuropeptide Y/drug effects , Receptors, Neuropeptide Y/physiology , Synapses/drug effects , Synapses/physiology
11.
Prog Neurobiol ; 196: 101894, 2021 01.
Article in English | MEDLINE | ID: mdl-32777329

ABSTRACT

An accelerating basic science literature is providing key insights into the mechanisms by which spinal neuropeptide Y (NPY) inhibits chronic pain. A key target of pain inhibition is the Gi-coupled neuropeptide Y1 receptor (Y1). Y1 is located in key sites of pain transmission, including the peptidergic subpopulation of primary afferent neurons and a dense subpopulation of small, excitatory, glutamatergic/somatostatinergic interneurons (Y1-INs) that are densely expressed in the dorsal horn, particularly in superficial lamina I-II. Selective ablation of spinal Y1-INs with an NPY-conjugated saporin neurotoxin attenuates the development of peripheral nerve injury-induced mechanical and cold hypersensitivity. Conversely, conditional knockdown of NPY expression or intrathecal administration of Y1 antagonists reinstates hypersensitivity in models of chronic latent pain sensitization. These and other results indicate that spinal NPY release and the consequent inhibition of pain facilitatory Y1-INs represent an important mechanism of endogenous analgesia. This mechanism can be mimicked with exogenous pharmacological approaches (e.g. intrathecal administration of Y1 agonists) to inhibit mechanical and thermal hypersensitivity and spinal neuron activity in rodent models of neuropathic, inflammatory, and postoperative pain. Pharmacological activation of Y1 also inhibits mechanical- and histamine-induced itch. These immunohistochemical, pharmacological, and cell type-directed lesioning data, in combination with recent transcriptomic findings, point to Y1-INs as a promising therapeutic target for the development of spinally directed NPY-Y1 agonists to treat both chronic pain and itch.


Subject(s)
Chronic Pain , Interneurons , Neuropeptide Y , Pruritus , Receptors, Neuropeptide Y , Spinal Cord , Animals , Chronic Pain/drug therapy , Chronic Pain/metabolism , Interneurons/drug effects , Interneurons/metabolism , Neuropeptide Y/drug effects , Neuropeptide Y/metabolism , Pruritus/drug therapy , Pruritus/metabolism , Receptors, Neuropeptide Y/drug effects , Receptors, Neuropeptide Y/metabolism , Spinal Cord/drug effects , Spinal Cord/metabolism
12.
J Neurochem ; 113(3): 615-27, 2010 May.
Article in English | MEDLINE | ID: mdl-20132466

ABSTRACT

We have shown that neuropeptide Y (NPY), a peptide neurotransmitter released by hippocampal interneurons, is proliferative for hippocampal neural stem progenitor cells (NSPCs) via the Y1 receptor. Fibroblast growth factor (FGF) 2, released predominantly by astrocytes, is also a powerful mitogen for postnatal and adult NSPCs, via the FGFR1 receptor. Knockout studies show that NPY and FGF2 are individually necessary, but not sufficient, for seizure-induced neurogenesis, suggesting a possible interaction. Here, we examined for interactions between NPY and FGF2 on NSPCs from the postnatal hippocampus and report that the combination of NPY and FGF2 significantly shortens the cell cycle time of nestin positive NSPCs, more than either factor alone. This augmentation of proliferation rate is NPY Y1 receptor mediated, and Y1 receptor activation increases both FGFR1 mRNA and protein in NSPC cultures. NSPCs immunostain for both Y1 and FGFR1 receptors and the interaction is specific for dentate NSPCs. This is the first report of a proliferative factor that augments the proliferative effect of FGF2 and is the first evidence of a positive proliferative interaction between a glial growth factor and a neuronal transmitter, identifying a novel neural activity driven mechanism for modulating the proliferation of hippocampal NSPCs.


Subject(s)
Fibroblast Growth Factor 2/pharmacology , Hippocampus/cytology , Hippocampus/metabolism , Intermediate Filament Proteins/biosynthesis , Nerve Tissue Proteins/biosynthesis , Neurons/metabolism , Neuropeptide Y/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/biosynthesis , Receptors, Neuropeptide Y/physiology , Stem Cells/metabolism , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Blotting, Western , Cell Proliferation/drug effects , Cells, Cultured , Homeodomain Proteins/metabolism , Image Processing, Computer-Assisted , Immunohistochemistry , Intermediate Filament Proteins/genetics , Nerve Tissue Proteins/genetics , Nestin , Neurons/drug effects , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Wistar , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptors, Neuropeptide Y/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/drug effects , Tumor Suppressor Proteins/metabolism
13.
Hippocampus ; 20(7): 820-8, 2010 Jul.
Article in English | MEDLINE | ID: mdl-19623606

ABSTRACT

One hypothesis of depression is that it is caused by reduced neuronal plasticity including hippocampal neurogenesis. In this study, we compared the effects of three long-term antidepressant treatments: escitalopram, voluntary running, and their combination on hippocampal cell proliferation, NPY and the NPY-Y1 receptor mRNAs, targets assumed to be important for hippocampal plasticity and mood disorders. An animal model of depression, the Flinders Sensitive Line (FSL) rat, was used and female rats were chosen because the majority of the depressed population is females. We investigated if these treatments were correlated to immobility, swimming, and climbing behaviors, which are associated with an overall, serotonergic-like and noradrenergic-like antidepressant response, in the Porsolt swim test (PST). Interestingly, while escitalopram, running and their combination increased the number of hippocampal BrdU immunoreactive cells, the antidepressant-like effect was only detected in the running group and the group with access both to running wheel and escitalopram. Hippocampal NPY mRNA and the NPY-Y1 receptor mRNA were elevated by running and the combined treatment. Moreover, correlations were detected between NPY mRNA levels and climbing and cell proliferation and NPY-Y1 receptor mRNA levels and swimming. Our results suggest that increased cell proliferation is not necessarily associated with an antidepressant effect. However, treatments that were associated with an antidepressant-like effect did regulate hippocampal levels of mRNAs encoding NPY and/or the NPY-Y1 receptor and support the notion that NPY can stimulate cell proliferation and induce an antidepressant-like response.


Subject(s)
Antidepressive Agents/pharmacology , Citalopram/pharmacology , Depression/physiopathology , Motor Activity/physiology , Neuropeptide Y/biosynthesis , Receptors, Neuropeptide Y/biosynthesis , Animals , Bromodeoxyuridine , Cell Proliferation , Depression/metabolism , Depression/therapy , Disease Models, Animal , Female , Hippocampus/metabolism , Hippocampus/physiopathology , Immunohistochemistry , In Situ Hybridization , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Neuropeptide Y/drug effects , RNA, Messenger/analysis , Rats , Receptors, Neuropeptide Y/drug effects , Running
14.
Diabetes Obes Metab ; 12(7): 591-603, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20590734

ABSTRACT

AIMS: Energy homeostasis is regulated by a complex interaction of molecules and pathways, and new antiobesity treatments are likely to require multiple pharmacological targeting of anorexigenic or orexigenic pathways to achieve effective loss of excess body weight and adiposity. Cannabinoids, acting via the cannabinoid-1 (CB1) receptor, and neuropeptide Y (NPY) are important modulators of feeding behaviour, energy metabolism and body composition. We investigated the interaction of CB1 and NPY in the regulation of energy homeostasis, hypothesizing that dual blockade of CB1 and NPY signalling will induce greater weight and/or fat loss than that induced by single blockade of either system alone. METHODS: We studied the effects of the CB1 antagonist Rimonabant on food intake, body weight, body composition, energy metabolism and bone physiology in wild-type (WT) and NPY knockout (NPY(-/-)) mice. Rimonabant was administered orally at 10 mg/kg body weight twice per day for 3 weeks. Oral Rimonabant was delivered voluntarily to mice via a novel method enabling studies to be carried out in the absence of gavage-induced stress. RESULTS: Mice with dual blockade of CB1 and NPY signalling (Rimonabant-treated NPY(-/-) mice) exhibited greater reductions in body weight and adiposity than mice with single blockade of either system alone (Rimonabant-treated WT or vehicle-treated NPY(-/-) mice). These changes occurred without loss of lean tissue mass or bone mass. Furthermore, Rimonabant-treated NPY(-/-) mice showed a lower respiratory exchange ratio than that seen in Rimonabant-treated WT or vehicle-treated NPY(-/-) mice, suggesting that this additive effect of dual blockade of CB1 and NPY involves promotion of lipid oxidation. On the other hand, energy expenditure and physical activity were comparable amongst all treatment groups. Interestingly, Rimonabant similarly and transiently reduced spontaneous and fasting-induced food intake in WT and NPY(-/-) mice in the first hour after administration only, suggesting independent regulation of feeding by CB1 and NPY signalling. In contrast, Rimonabant increased serum corticosterone levels in WT mice, but this effect was not seen in NPY(-/-) mice, indicating that NPY signalling may be required for effects of CB1 on the hypothalamo-pituitary-adrenal axis. CONCLUSIONS: Dual blockade of CB1 and NPY signalling leads to additive reductions in body weight and adiposity without concomitant loss of lean body mass or bone mass. An additive increase in lipid oxidation in dual CB1 and NPY blockade may contribute to the effect on adiposity. These findings open new avenues for more effective treatment of obesity via dual pharmacological manipulations of the CB1 and NPY systems.


Subject(s)
Eating/drug effects , Neuropeptide Y/drug effects , Obesity/drug therapy , Piperidines/antagonists & inhibitors , Pyrazoles/antagonists & inhibitors , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptors, Neuropeptide Y/drug effects , Animals , Body Composition , Energy Metabolism/drug effects , Homeostasis/physiology , Mice , Neuropeptide Y/metabolism , Obesity/metabolism , Oxidation-Reduction , Piperidines/administration & dosage , Pyrazoles/administration & dosage , Receptor, Cannabinoid, CB1/administration & dosage , Rimonabant
15.
Int Endod J ; 43(10): 835-42, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20636351

ABSTRACT

AIM: To investigate whether dental pulp fibroblasts express neuropeptide Y (NPY) and NPY-Y1 in vitro and to determine the effects of the cytokines including interleukin-1ß (IL-1ß), TGF- ß(1) , substance P and NPY on the expression of NPY Y1. METHODOLOGY: Three primary fibroblast cell strains were obtained from freshly extracted human third molar teeth. RT-PCR was utilized to detect expression of NPY and mRNA expression. Membrane protein samples were isolated, and protein expression was determined by Western blotting. Radioimmunoassay was used to quantify NPY expression in healthy (n = 35) and carious (n = 39) whole pulp samples, and the student's t-test was used to test for statistical significance. In addition, the 3-(4,5-Dimethylthiazol,2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay was used to assay fibroblast cell growth. RESULTS: mRNA transcripts were found in all three fibroblast cell populations with the cytokines having a stimulatory effect on its expression (P < 0.05). NPY mRNA was not detected in the cell strains. NPY-Y1 receptor protein expression was visualized by Western blotting, and there was no effect of IL-1ß or TGF- ß(1) on its expression. The mean concentration of NPY-Ir determined by radioimmunoassay in non-carious teeth was 19.40 ng x g(-1) (±17.03 SD) compared to 29.95 ng x g(-1) (±20.99 SD) in carious teeth (P < 0.05). CONCLUSION: Human dental pulp fibroblasts express, but do not synthesize, NPY, demonstrating that the fibroblast is a target cell for NPY. The effect of proinflammatory cytokines suggests that fibroblasts play a neuroimmunomodulatory role in the pulpal response to dental caries and injury.


Subject(s)
Dental Pulp/metabolism , Fibroblasts/metabolism , Neuropeptide Y/analysis , Receptors, Neuropeptide Y/analysis , Blotting, Western , Cell Culture Techniques , Cell Proliferation/drug effects , Cells, Cultured , Coloring Agents , Dental Caries/metabolism , Dental Pulp/cytology , Dental Pulp/drug effects , Fibroblasts/drug effects , Humans , Interleukin-1beta/pharmacology , Membrane Proteins/analysis , Neuroimmunomodulation , Neuropeptide Y/pharmacology , Receptors, Neuropeptide Y/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Substance P/pharmacology , Tetrazolium Salts , Thiazoles , Transforming Growth Factor beta/pharmacology
16.
Neuropeptides ; 80: 102001, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31916978

ABSTRACT

The neuropeptide Y (NPY) system plays an important role in mediating resilience to the harmful effect of stress in post-traumatic stress disorder (PTSD). It can mediate its effects via several G-protein coupled receptors: Y1R, Y2R, Y4R and Y5R. To investigate the role of individual NPY receptors in the resilience effects of NPY to traumatic stress, intranasal infusion of either Y1R agonists [D-His26]NPY, [Leu31Pro34]NPY, Y2R agonist NPY (3-36) or NPY were administered to male Sprague-Dawley rats immediately following the last stressor of the single prolonged stress (SPS) protocol, a widely used PTSD animal model. After 7 or 14 days, effects of the treatments were measured on the elevated plus maze (EPM) for anxiety, in forced swim test (FST) for development of depressive-like or re-experiencing behavior, in social interaction (SI) test for impaired social behavior, and acoustic startle response (ASR) for hyperarousal. [D-His26]NPY, but not [Leu31Pro34]NPY nor NPY (3-36) Y2R, was effective in preventing the SPS-elicited development of anxiety. Y1R, but not Y2R agonists prevented development of depressive- feature on FST, with [D-His26]NPY superior to NPY. The results demonstrate that [D-His26]NPY was sufficient to prevent development of anxiety, social impairment and depressive symptoms, and has promise as an early intervention therapy following traumatic stress.


Subject(s)
Anxiety/drug therapy , Neuropeptide Y/pharmacology , Receptors, Neuropeptide Y/drug effects , Reflex, Startle/drug effects , Administration, Intranasal , Animals , Anxiety/chemically induced , Disease Models, Animal , Male , Maze Learning/drug effects , Rats, Sprague-Dawley , Stress Disorders, Post-Traumatic/chemically induced , Stress Disorders, Post-Traumatic/drug therapy
17.
Neuropeptides ; 80: 102029, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32127176

ABSTRACT

Differentiation, self-renewal and quiescence of Hematopoietic stem cells (HSCs) is tightly regulated in order to protect the HSCs from the strain of constant cell division and depletion of the stem cell pool. The neurotransmitter Neuropeptide Y (NPY) is released from sympathetic nerves in the bone marrow and has been shown to indirectly affect HSC function through effects on bone marrow (BM) multipotent Mesenchymal Stromal Cells (MSCs), osteoblasts (OBs) and macrophages. Although the absence of NPY has been shown to be accompanied by severe BM impairment and delayed engraftment of HSCs, the direct effects of NPY on HSCs have never been assessed. Here, we aimed to explore the effect of NPY on the regulation of HSCs. All NPY receptors Y1, Y2, Y4 and Y5 were found to be highly expressed on most HSCs and mature hematopoietic cell subsets. In culture, in particularly expression of the Y1 receptor was shown to decrease in time. Doses of 300 nM NPY suppressed HSC proliferation in cell cultures, as confirmed by an increase of HSCs in G0 phase and an increase in the gene expression levels of FOXO3, DICER1, SMARCA2 and PDK1, which all have been shown to play an important role in the regulation of cell quiescence. These data support the idea that NPY may have a direct effect on the regulation of HSC fate by modulating cell quiescence.


Subject(s)
Cell Proliferation/drug effects , Hematopoietic Stem Cells/drug effects , Neuropeptide Y/pharmacology , Receptors, Neuropeptide Y/metabolism , Cell Differentiation/drug effects , Cell Division/drug effects , Cells, Cultured , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Neuropeptide Y/metabolism , Receptors, Neuropeptide Y/drug effects
18.
Eur Neuropsychopharmacol ; 33: 126-138, 2020 04.
Article in English | MEDLINE | ID: mdl-32057593

ABSTRACT

The adolescent brain displays high vulnerability to the deleterious effects of ethanol, including greater risk of developing alcohol use disorder later in life. Here, we characterized the gene expression of the endocannabinoid system (ECS) and relevant signaling systems associated with neuroinflammation and emotional behaviors in the brain of young adult control and ethanol-exposed (EtOH) rats. We measured mRNA levels of candidate genes using quantitative real time PCR in the medial prefrontal cortex (mPFC), amygdala and hippocampus. EtOH rats were generated by maintenance on an intermittent and voluntary ethanol consumption during adolescence using the two-bottle choice paradigm (4 days/week for 4 weeks) followed by 2 week-withdrawal, a time-point of withdrawal with no physical symptoms. Mean differences and effect sizes were calculated using t-test and Cohen's d values. In the mPFC and hippocampus, EtOH rats had significantly higher mRNA expression of endocannabinoid-signaling (mPFC: Ppara, Dagla, Daglb and Napepld; and hippocampus: Cnr2, Dagla and Mgll) and neuroinflammation-associated genes (mPFC: Gfap; and hippocampus: Aif1) than in controls. Moreover, EtOH rats had significantly higher mRNA expression of neuropeptide Y receptor genes (Npy1r, Npy2r and Npy5r) in the hippocampus. Finally, EtOH rats also displayed higher plasma endocannabinoid levels than controls. In conclusion, these results suggest that adolescent ethanol exposure can lead to long-term alterations in the gene expression of the ECS and other signaling systems involved in neuroinflammation and regulation of emotional behaviors in key brain areas for the development of addiction.


Subject(s)
Alcohol Drinking/adverse effects , Central Nervous System Depressants/adverse effects , Endocannabinoids/genetics , Endocannabinoids/metabolism , Ethanol/adverse effects , Inflammation Mediators/metabolism , Animals , Anxiety/psychology , Emotions , Gene Expression/drug effects , Male , Memory/drug effects , Motor Activity/drug effects , Psychology, Adolescent , Psychomotor Performance/drug effects , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Wistar , Receptors, Neuropeptide Y/drug effects , Receptors, Neuropeptide Y/genetics , Receptors, Neuropeptide Y/metabolism
19.
Neuropeptides ; 79: 101979, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31708112

ABSTRACT

Neuropeptide Y (NPY) is an endogenous neuropeptide that is abundantly expressed in the central nervous system. NPY is involved in various neurological processes and neuropsychiatric disorders, including fear learning and anxiety disorders. Reduced levels of NPY are reported in Post-Traumatic Stress Disorder (PTSD) patients, and NPY has been proposed as a potential therapeutic target for PTSD. It is therefore important to understand the effects of chronic enhancement of NPY on anxiety and fear learning. Previous studies have shown that acute elevation of NPY reduces anxiety, fear learning and locomotor activity. Models of chronic NPY overexpression have produced mixed results, possibly caused by ectopic NPY expression. NPY is expressed primarily by a subset of GABAergic interneurons, providing specific spatiotemporal release patterns. Administration of exogenous NPY throughout the brain, or overexpression in cells that do not normally release NPY, can have detrimental side effects, including memory impairment. In order to determine the effects of boosting NPY only in the cells that normally release it, we utilized a transgenic mouse line that overexpresses NPY only in NPY+ cells. We tested for effects on anxiety related behaviors in adolescent mice, an age with high incidence of anxiety disorders in humans. Surprisingly, we did not observe the expected reduction in anxiety-like behavior in NPY overexpression mice. There was no change in fear learning behavior, although there was a deficit in nest building. The effect of exogenous NPY on synaptic transmission in acute hippocampal slices was also diminished, indicating that the function of NPY receptors is impaired. Reduced NPY receptor function could contribute to the unexpected behavioral outcomes. We conclude that overexpression of NPY, even in cells that normally express it, can lead to reduced responsiveness of NPY receptors, potentially affecting the ability of NPY to function as a long-term therapeutic.


Subject(s)
Anxiety/metabolism , Brain/metabolism , Neuropeptide Y/metabolism , Receptors, Neuropeptide Y/metabolism , Stress Disorders, Post-Traumatic/metabolism , Animals , Brain/drug effects , Hippocampus/metabolism , Maze Learning/drug effects , Mice, Transgenic , Neuropeptide Y/pharmacology , Neuropeptides/metabolism , Receptors, Neuropeptide Y/drug effects , Stress Disorders, Post-Traumatic/drug therapy , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
20.
Neuropeptides ; 80: 102024, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32145934

ABSTRACT

Transection of the sural and common peroneal branches of the sciatic nerve produces cutaneous hypersensitivity at the tibial innervation territory of the mouse hindpaw that resolves within a few weeks. We report that interruption of endogenous neuropeptide Y (NPY) signaling during remission, with either conditional NPY knockdown in NPYtet/tet mice or intrathecal administration of the Y1 receptor antagonist BIBO3304, reinstated hypersensitivity. These data indicate that nerve injury establishes a long-lasting latent sensitization of spinal nociceptive neurons that is masked by spinal NPY-Y1 neurotransmission. To determine whether this mechanism extends beyond the sensory component of nociception, we used conditioned place aversion and preference assays to evaluate the affective component of pain. We found that BIBO3304 produced place aversion in mice when administered during remission. Furthermore, the analgesic drug gabapentin produced place preference after NPY knockdown in NPYtet/tet but not control mice. We then used pharmacological agents and deletion mutant mice to investigate the cellular mechanisms of neuropathic latent sensitization. BIBO3304-induced reinstatement of mechanical hypersensitivity and conditioned place aversion could be prevented with intrathecal administration of an N-methyl-d-aspartate receptor antagonist (MK-801) and was absent in adenylyl cyclase type 1 (AC1) deletion mutant mice. BIBO3304-induced reinstatement could also be prevented with intrathecal administration an AC1 inhibitor (NB001) or a TRPV1 channel blocker (AMG9801), but not vehicle. Intrathecal administration of a TRPA1 channel blocker (HC030031) prevented the reinstatement of neuropathic hypersensitivity produced either by BIBO3304, or by NPY knockdown in NPYtet/tet but not control mice. Our results confirm new mediators of latent sensitization: TRPA1 and TRPV1. We conclude that NPY acts at spinal Y1 to tonically inhibit a molecular NMDAR➔AC1 intracellular signaling pathway in the dorsal horn that is induced by peripheral nerve injury and drives both the sensory and affective components of chronic neuropathic pain.


Subject(s)
Adenylyl Cyclases/pharmacology , Hyperalgesia/drug therapy , Neuropeptide Y/pharmacology , Pain/drug therapy , Adenylyl Cyclases/metabolism , Analgesics/pharmacology , Animals , Hyperalgesia/metabolism , Male , Mice , Neuralgia/metabolism , Neuropeptide Y/metabolism , Nociception/drug effects , Peripheral Nerve Injuries/drug therapy , Peripheral Nerve Injuries/metabolism , Posterior Horn Cells/drug effects , Posterior Horn Cells/metabolism , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Neuropeptide Y/drug effects , Receptors, Neuropeptide Y/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL