Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.639
Filter
1.
Cell Rep Med ; 5(5): 101559, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38744275

ABSTRACT

Dysfunction of the sympathetic nervous system and increased epicardial adipose tissue (EAT) have been independently associated with the occurrence of cardiac arrhythmia. However, their exact roles in triggering arrhythmia remain elusive. Here, using an in vitro coculture system with sympathetic neurons, cardiomyocytes, and adipocytes, we show that adipocyte-derived leptin activates sympathetic neurons and increases the release of neuropeptide Y (NPY), which in turn triggers arrhythmia in cardiomyocytes by interacting with the Y1 receptor (Y1R) and subsequently enhancing the activity of the Na+/Ca2+ exchanger (NCX) and calcium/calmodulin-dependent protein kinase II (CaMKII). The arrhythmic phenotype can be partially blocked by a leptin neutralizing antibody or an inhibitor of Y1R, NCX, or CaMKII. Moreover, increased EAT thickness and leptin/NPY blood levels are detected in atrial fibrillation patients compared with the control group. Our study provides robust evidence that the adipose-neural axis contributes to arrhythmogenesis and represents a potential target for treating arrhythmia.


Subject(s)
Adipocytes , Adipose Tissue , Arrhythmias, Cardiac , Leptin , Myocytes, Cardiac , Neuropeptide Y , Pericardium , Humans , Animals , Pericardium/metabolism , Pericardium/pathology , Adipose Tissue/metabolism , Adipose Tissue/pathology , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Neuropeptide Y/metabolism , Leptin/metabolism , Adipocytes/metabolism , Male , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Neurons/metabolism , Neurons/pathology , Sodium-Calcium Exchanger/metabolism , Female , Receptors, Neuropeptide Y/metabolism , Middle Aged , Atrial Fibrillation/metabolism , Atrial Fibrillation/physiopathology , Atrial Fibrillation/pathology , Sympathetic Nervous System/metabolism , Mice , Epicardial Adipose Tissue
2.
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
3.
Cells ; 13(8)2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38667284

ABSTRACT

This study investigates the combined effects of the neuropeptide Y Y1 receptor (NPY1R) agonist [Leu31-Pro34]NPY at a dose of 132 µg and Ketamine at 10 mg/Kg on cognitive functions and neuronal proliferation, against a backdrop where neurodegenerative diseases present an escalating challenge to global health systems. Utilizing male Sprague-Dawley rats in a physiological model, this research employed a single-dose administration of these compounds and assessed their impact 24 h after treatment on object-in-place memory tasks, alongside cellular proliferation within the dorsal hippocampus dentate gyrus. Methods such as the in situ proximity ligation assay and immunohistochemistry for proliferating a cell nuclear antigen (PCNA) and doublecortin (DCX) were utilized. The results demonstrated that co-administration significantly enhanced memory consolidation and increased neuronal proliferation, specifically neuroblasts, without affecting quiescent neural progenitors and astrocytes. These effects were mediated by the potential formation of NPY1R-TrkB heteroreceptor complexes, as suggested by receptor co-localization studies, although further investigation is required to conclusively prove this interaction. The findings also highlighted the pivotal role of brain-derived neurotrophic factor (BDNF) in mediating these effects. In conclusion, this study presents a promising avenue for enhancing cognitive functions and neuronal proliferation through the synergistic action of the NPY1R agonist and Ketamine, potentially via NPY1R-TrkB heteroreceptor complex formation, offering new insights into therapeutic strategies for neurodegenerative diseases.


Subject(s)
Cell Proliferation , Cognition , Doublecortin Protein , Ketamine , Neurons , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled , Receptors, Neuropeptide Y , Receptors, Neuropeptide , Animals , Male , Ketamine/pharmacology , Ketamine/administration & dosage , Cognition/drug effects , Rats , Receptors, Neuropeptide Y/agonists , Receptors, Neuropeptide Y/metabolism , Neurons/drug effects , Neurons/metabolism , Cell Proliferation/drug effects , Receptor, trkB/agonists , Receptor, trkB/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Dentate Gyrus/drug effects , Dentate Gyrus/metabolism , Neurogenesis/drug effects
4.
Acta Med Okayama ; 78(2): 95-106, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38688827

ABSTRACT

The lungs are very complex organs, and the respiratory system performs the dual roles of repairing tissue while protecting against infection from various environmental stimuli. Persistent external irritation disrupts the immune responses of tissues and cells in the respiratory system, ultimately leading to respiratory disease. Neuropeptide Y (NPY) is a 36-amino-acid polypeptide and a neurotransmitter that regulates homeostasis. The NPY receptor is a seven-transmembrane-domain G-protein-coupled receptor with six subtypes (Y1, Y2, Y3, Y4, Y5, and Y6). Of these receptors, Y1, Y2, Y4, and Y5 are functional in humans, and Y1 plays important roles in the immune responses of many organs, including the respiratory system. NPY and the Y1 receptor have critical roles in the pathogenesis of asthma, chronic obstructive pulmonary disease, and idiopathic pulmonary fibrosis. The effects of NPY on the airway immune response and pathogenesis differ among respiratory diseases. This review focuses on the involvement of NPY in the airway immune response and pathogenesis of various respiratory diseases.


Subject(s)
Neuropeptide Y , Receptors, Neuropeptide Y , Humans , Neuropeptide Y/physiology , Neuropeptide Y/metabolism , Receptors, Neuropeptide Y/physiology , Animals , Respiratory Tract Diseases/immunology , Asthma/immunology , Respiratory System/immunology , Pulmonary Disease, Chronic Obstructive/immunology
5.
Expert Opin Ther Targets ; 28(4): 295-308, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38622072

ABSTRACT

BACKGROUND: Major Depressive Disorder (MDD) is a prevalent and debilitating condition, necessitating novel therapeutic strategies due to the limited efficacy and adverse effects of current treatments. We explored how galanin receptor 2 (GALR2) and Neuropeptide Y1 Receptor (NPYY1R) agonists, working together, can boost brain cell growth and increase antidepressant-like effects in rats. This suggests new ways to treat Major Depressive Disorder (MDD). RESEARCH DESIGN AND METHODS: In a controlled laboratory setting, adult naive Sprague-Dawley rats were administered directly into the brain's ventricles, a method known as intracerebroventricular (ICV) administration, with GALR2 agonist (M1145), NPYY1R agonist, both, or in combination with a GALR2 antagonist (M871). Main outcome measures included long-term neuronal survival, differentiation, and behavioral. RESULTS: Co-administration of M1145 and NPYY1R agonist significantly enhanced neuronal survival and maturation in the ventral dentate gyrus, with a notable increase in Brain-Derived Neurotrophic Factor (BDNF) expression. This neurogenic effect was associated with an antidepressant-like effect, an outcome partially reversed by M871. CONCLUSIONS: GALR2 and NPYY1R agonists jointly promote hippocampal neurogenesis and exert antidepressant-like effects in rats without adverse outcomes, highlighting their therapeutic potential for MDD. The study's reliance on an animal model and intracerebroventricular delivery warrants further clinical exploration to confirm these promising results.


Subject(s)
Antidepressive Agents , Brain-Derived Neurotrophic Factor , Cell Survival , Depressive Disorder, Major , Neurons , Rats, Sprague-Dawley , Receptor, Galanin, Type 2 , Receptors, Neuropeptide Y , Animals , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/physiopathology , Receptor, Galanin, Type 2/metabolism , Rats , Brain-Derived Neurotrophic Factor/metabolism , Male , Receptors, Neuropeptide Y/metabolism , Receptors, Neuropeptide Y/antagonists & inhibitors , Neurons/drug effects , Neurons/metabolism , Cell Survival/drug effects , Antidepressive Agents/pharmacology , Antidepressive Agents/administration & dosage , Disease Models, Animal , Peptides , Receptors, Neuropeptide , Receptors, G-Protein-Coupled
6.
Expert Opin Ther Targets ; 28(4): 309-322, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38626283

ABSTRACT

BACKGROUND: Major Depressive Disorder (MDD) poses a significant challenge to global health, with current treatments often limited by efficacy and onset delays. This study explores the synergistic antidepressant-like effects of an NPY1R agonist and Ketamine, targeting their neurobiological interactions within the ventral hippocampus. RESEARCH DESIGN AND METHODS: Utilizing a preclinical model, this study administered Neuropeptide Y receptor 1 (NPY1R) agonist and Ketamine, both separately and in combination, through intracerebroventricular (icv) and intranasal (i.n.) routes. The Forced Swimming Test (FST) was employed to assess antidepressant-like activity, while in situ Proximity Ligation Assay and immunohistochemistry were used to examine NPY1R/TrkB heteroreceptor complexes and BDNF expression in the ventral dentate gyrus (DG), along with neurogenesis markers. RESULTS: The combined treatment significantly reduced immobility in the FST, indicative of enhanced antidepressant-like effects, correlated with increased formation of NPY1R/TrkB complex and brain-derived neurotrophic factor (BDNF) expression in the ventral DG. These molecular alterations were associated with increased neurogenesis. CONCLUSIONS: The coadministration of an NPY1R agonist and Ketamine in a rodent model demonstrated potentiated antidepressant responses through synergistic neurobiological pathways, including TrkB signaling and hippocampal neurogenesis. This indicates a novel therapeutic strategy for MDD, warranting further clinical investigation to fully understand its implications.


Subject(s)
Antidepressive Agents , Brain-Derived Neurotrophic Factor , Depressive Disorder, Major , Drug Synergism , Hippocampus , Ketamine , Neurogenesis , Receptor, trkB , Receptors, Neuropeptide Y , Signal Transduction , Animals , Neurogenesis/drug effects , Antidepressive Agents/pharmacology , Antidepressive Agents/administration & dosage , Male , Brain-Derived Neurotrophic Factor/metabolism , Signal Transduction/drug effects , Receptors, Neuropeptide Y/agonists , Receptors, Neuropeptide Y/metabolism , Depressive Disorder, Major/drug therapy , Ketamine/pharmacology , Ketamine/administration & dosage , Hippocampus/metabolism , Hippocampus/drug effects , Receptor, trkB/agonists , Receptor, trkB/metabolism , Disease Models, Animal , Rats , Mice , Rats, Sprague-Dawley , Dentate Gyrus/drug effects , Dentate Gyrus/metabolism , Swimming
7.
FASEB J ; 38(7): e23595, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38572811

ABSTRACT

This study evaluates the sustained antidepressant-like effects and neurogenic potential of a 3-day intranasal co-administration regimen of galanin receptor 2 (GALR2) agonist M1145 and neuropeptide Y Y1 receptor (NPY1R) agonist [Leu31, Pro34]NPY in the ventral hippocampus of adult rats, with outcomes analyzed 3 weeks post-treatment. Utilizing the forced swimming test (FST), we found that this co-administration significantly enhances antidepressant-like behaviors, an effect neutralized by the GALR2 antagonist M871, highlighting the synergistic potential of these neuropeptides in modulating mood-related behaviors. In situ proximity ligation assay (PLA) indicated a significant increase in GALR2/NPYY1R heteroreceptor complexes in the ventral hippocampal dentate gyrus, suggesting a molecular basis for the behavioral outcomes observed. Moreover, proliferating cell nuclear antigen (PCNA) immunolabeling revealed increased cell proliferation in the subgranular zone of the dentate gyrus, specifically in neuroblasts as evidenced by co-labeling with doublecortin (DCX), without affecting quiescent neural progenitors or astrocytes. The study also noted a significant uptick in the number of DCX-positive cells and alterations in dendritic morphology in the ventral hippocampus, indicative of enhanced neuronal differentiation and maturation. These morphological changes highlight the potential of these agonists to facilitate the functional integration of new neurons into existing neural circuits. By demonstrating the long-lasting effects of a brief, 3-day intranasal administration of GALR2 and NPY1R agonists, our findings contribute significantly to the understanding of neuropeptide-mediated neuroplasticity and herald novel therapeutic strategies for the treatment of depression and related mood disorders, emphasizing the therapeutic promise of targeting neurogenesis and neuronal maturation processes.


Subject(s)
Neuropeptide Y , Neuropeptides , Rats , Animals , Receptor, Galanin, Type 2/agonists , Receptor, Galanin, Type 2/metabolism , Administration, Intranasal , Galanin/pharmacology , Galanin/metabolism , Hippocampus/metabolism , Receptors, Neuropeptide Y/metabolism , Neuropeptides/pharmacology , Antidepressive Agents/pharmacology , Neurogenesis
8.
Neurosci Lett ; 825: 137707, 2024 Mar 10.
Article in English | MEDLINE | ID: mdl-38431039

ABSTRACT

Visfatin play an essential role in the central regulation of appetite in birds. This study aimed to determine role of intracerebroventricular (ICV) injection of the visfatin on food intake and its possible interaction with neuropeptide Y (NPY) and nitric oxide system in neonatal broiler chicken. In experiment 1, neonatal chicken received ICV injection visfatin (1, 2 and 4 µg). In experiment 2, chicken received ICV injection of B5063 (NPY1 receptor antagonist 1.25 µg), visfatin (4 µg) and co-injection of the B5063 + Visfatin. In experiments 3-6, SF22 (NPY2 receptor antagonist 1.25 µg), SML0891 (NPY5 receptor antagonist 1.25 µg), L-NAME (nitric oxide synthase inhibitor, 100 nmol) and L-arginine (Precursor of nitric oxide, 200 nmol) were injected instead of B5063. Then the amount of cumulative food was measured at 30, 60 and 120 min after injection. Obtained data showed, injection visfatin (2 and 4 µg) increased food intake compared to control group (P < 0.05). Co-injection of the B5063 + Visfatin decreased visfatin-induced hyperphagia compared to control group (P < 0.05). Co-injection of the L-NAME + Visfatin amplified visfatin-induced hyperphagia compared to control group (P < 0.05). The result showed that visfatin has hyperphagic role and this effect mediates via NPY1 and nitric oxide system in neonatal chicken.


Subject(s)
Chickens , Neuropeptide Y , Animals , Animals, Newborn , Neuropeptide Y/pharmacology , Chickens/physiology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide , Nicotinamide Phosphoribosyltransferase , Eating , Receptors, Neuropeptide Y , Hyperphagia , Feeding Behavior/physiology
9.
Int J Mol Sci ; 25(6)2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38542321

ABSTRACT

Our objective was to investigate the effects of topically applied neuropeptide Y (NPY) on ischemic wounds. Initially, the animal model for ischemic wound healing was validated using 16 male Sprague Dawley albino rats. In the intervention study, an additional 28 rats were divided into three groups: NPY (0.025%), the positive control insulin-like growth factor-I (IGF-I, 0.0025%), and the hydrogel carrier alone (control). The hydrogel was selected due to its capacity to prolong NPY release (p < 0.001), as demonstrated in a Franz diffusion cell. In the animals, an 8 mm full-thickness wound was made in a pedunculated dorsal ischemic skin flap. Wounds were then treated and assessed for 14 days and collected at the end of the experiment for in situ hybridization analysis (RNAscope®) targeting NPY receptor Y2R and for meticulous histologic examination. Wound healing rates, specifically the percentage changes in wound area, did not show an increase with NPY (p = 0.907), but there was an increase with rhIGF-I (p = 0.039) compared to the control. Y2R mRNA was not detected in the wounds or adjacent skin but was identified in the rat brain (used as a positive control). Light microscopic examination revealed trends of increased angiogenesis and enhanced inflammatory cell infiltration with NPY compared to control. An interesting secondary discovery was the presence of melanophages in the wounds. Our findings suggest the potential of NPY to enhance neovascularization under ischemic wound healing conditions, but further optimization of the carrier and dosage is necessary. The mechanism remains elusive but likely involves NPY receptor subtypes other than Y2R.


Subject(s)
Neuropeptide Y , Wound Healing , Rats , Male , Animals , Neuropeptide Y/genetics , Neuropeptide Y/pharmacology , Rats, Sprague-Dawley , Receptors, Neuropeptide Y , Hydrogels/pharmacology
10.
Nutrients ; 16(6)2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38542814

ABSTRACT

(1) Background: Modulators of the Neuropeptide Y (NPY) system are involved in energy metabolism, but the effect of NPY receptor antagonists on metabolic-dysfunction-associated steatotic liver disease (MASLD), a common obesity-related comorbidity, are largely unknown. In this study, we report on the effects of antagonists of the NPY-2 receptor (Y2R) in comparison with empagliflozin and semaglutide, substances that are known to be beneficial in MASLD. (2) Methods: Diet-induced obese (DIO) male Wistar rats were randomized into the following treatment groups: empagliflozin, semaglutide ± PYY3-36, the Y2R antagonists JNJ 31020028 and a food-restricted group, as well as a control group. After a treatment period of 8 weeks, livers were weighed and histologically evaluated. QrtPCR was performed to investigate liver inflammation and de novo lipogenesis (in liver and adipose tissue). Serum samples were analysed for metabolic parameters. (3) Results: Semaglutide + PYY3-36 led to significant weight loss, reduced liver steatosis (p = 0.05), and decreased inflammation, insulin resistance, and leptin levels. JNJ-31020028 prevented steatosis (p = 0.03) without significant weight loss. Hepatic downregulation of de novo lipogenesis-regulating genes (SREBP1 and MLXIPL) was observed in JNJ-31020028-treated rats (p ≤ 0.0001). Food restriction also resulted in significantly reduced weight, steatosis, and hepatic de novo lipogenesis. (4) Conclusions: Body weight reduction (e.g., by food restriction or drugs like semaglutide ± PYY3-36) is effective in improving liver steatosis in DIO rats. Remarkably, the body-weight-neutral Y2R antagonists may be effective in preventing liver steatosis through a reduction in de novo lipogenesis, making this drug class a candidate for the treatment of (early) MASLD.


Subject(s)
Benzamides , Benzhydryl Compounds , Fatty Liver , Glucagon-Like Peptides , Glucosides , Piperazines , Receptors, Neuropeptide Y , Rats , Male , Animals , Receptors, Neuropeptide Y/metabolism , Rats, Wistar , Obesity/complications , Obesity/drug therapy , Diet , Fatty Liver/drug therapy , Fatty Liver/etiology , Fatty Liver/prevention & control , Weight Loss , Inflammation
11.
Mol Pain ; 20: 17448069241242982, 2024.
Article in English | MEDLINE | ID: mdl-38485252

ABSTRACT

Itch is a somatosensory sensation to remove potential harmful stimulation with a scratching desire, which could be divided into mechanical and chemical itch according to diverse stimuli, such as wool fiber and insect biting. It has been reported that neuropeptide Y (NPY) neurons, a population of spinal inhibitory interneurons, could gate the transmission of mechanical itch, with no effect on chemical itch. In our study, we verified that chemogenetic activation of NPY neurons could inhibit the mechanical itch as well as the chemical itch, which also attenuated the alloknesis phenomenon in the chronic dry skin model. Afterwards, intrathecal administration of NPY1R agonist, [Leu31, Pro34]-NPY (LP-NPY), showed the similar inhibition effect on mechanical itch, chemical itch and alloknesis as chemo-activation of NPY neurons. Whereas, intrathecal administration of NPY1R antagonist BIBO 3304 enhanced mechanical itch and reversed the alloknesis phenomenon inhibited by LP-NPY treatment. Moreover, selectively knocking down NPY1R by intrathecal injection of Npy1r siRNA enhanced mechanical and chemical itch behavior as well. These results indicate that NPY neurons in spinal cord regulate mechanical and chemical itch, and alloknesis in dry skin model through NPY1 receptors.


Subject(s)
Neuropeptide Y , Receptors, Neuropeptide Y , Animals , Pruritus/chemically induced , Signal Transduction , Spinal Cord
12.
Behav Brain Funct ; 20(1): 6, 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38549164

ABSTRACT

BACKGROUND: Spatial memory deficits and reduced neuronal survival contribute to cognitive decline seen in the aging process. Current treatments are limited, emphasizing the need for innovative therapeutic strategies. This research explored the combined effects of intranasally co-administered galanin receptor 2 (GALR2) and neuropeptide Y1 receptor (NPY1R) agonists, recognized for their neural benefits, on spatial memory, neuronal survival, and differentiation in adult rats. After intranasal co-delivery of the GALR2 agonist M1145 and a NPY1R agonist to adult rats, spatial memory was tested with the object-in-place task 3 weeks later. We examined neuronal survival and differentiation by assessing BrdU-IR profiles and doublecortin (DCX) labeled cells, respectively. We also used the GALR2 antagonist M871 to confirm GALR2's crucial role in promoting cell growth. RESULTS: Co-administration improved spatial memory and increased the survival rate of mature neurons. The positive effect of GALR2 in cell proliferation was confirmed by the nullifying effects of its antagonist. The treatment boosted DCX-labeled newborn neurons and altered dendritic morphology, increasing cells with mature dendrites. CONCLUSIONS: Our results show that intranasal co-delivery of GALR2 and NPY1R agonists improves spatial memory, boosts neuronal survival, and influences neuronal differentiation in adult rats. The significant role of GALR2 is emphasized, suggesting new potential therapeutic strategies for cognitive decline.


Subject(s)
Cognitive Dysfunction , Receptor, Galanin, Type 2 , Rats , Animals , Receptor, Galanin, Type 2/agonists , Receptor, Galanin, Type 2/physiology , Receptors, Neuropeptide Y , Galanin/pharmacology , Neurogenesis , Cognition , Cognitive Dysfunction/drug therapy
13.
Neuropeptides ; 105: 102416, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38430725

ABSTRACT

The neuropeptide Y4 receptor (Y4R), a rhodopsin-like G protein-coupled receptor (GPCR) and the hormone pancreatic polypeptide (PP) are members of the neuropeptide Y family consisting of four receptors (Y1R, Y2R, Y4R, Y5R) and three highly homologous peptide ligands (neuropeptide Y, peptide YY, PP). In this family, the Y4R is of particular interest as it is the only subtype with high affinity to PP over NPY. The Y4R, as a mediator of PP signaling, has a pivotal role in appetite regulation and energy homeostasis, offering potential avenues for the treatment of metabolic disorders such as obesity. PP as anorexigenic peptide is released postprandial from the pancreas in response to food intake, induces satiety signals and contributes to hamper excessive food intake. Moreover, this system was also described to be associated with different types of cancer: overexpression of Y4R have been found in human adenocarcinoma cells, while elevated levels of PP are related to the development of pancreatic endocrine tumors. The pharmacological relevance of the Y4R advanced the search for potent and selective ligands for this receptor subtype, which will be significantly progressed through the elucidation of the active state PP-Y4R cryo-EM structure. This review summarizes the development of novel PP-derived ligands, like Obinepitide as dual Y2R/Y4R agonist in clinical trials or UR-AK86c as small hexapeptide agonist with picomolar affinity, as well as the first allosteric modulators that selectively target the Y4R, e.g. VU0506013 as potent Y4R positive allosteric modulator or (S)-VU0637120 as allosteric antagonist. Here, we provide valuable insights into the complex physiological functions of the Y4R and PP and the pharmacological relevance of the system in appetite regulation to open up new avenues for the development of tool compounds for targeted therapies with potential applications in metabolic disorders.


Subject(s)
Pancreatic Polypeptide , Receptors, Neuropeptide Y , Humans , Pancreatic Polypeptide/metabolism , Receptors, Neuropeptide Y/metabolism , Animals , Ligands
14.
Neuropeptides ; 105: 102425, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38554699

ABSTRACT

The control of feeding and physical activity is tightly linked and coordinated. However the underlying mechanisms are unclear. One of the major regulatory systems of feeding behaviour involves neuropeptide Y (NPY) signalling, with the signalling mediated through NPY Y4 receptor also known to influence activity. Here we show that mice globally lacking the Npy4r (Npy4r-/-) in the absence of access to a running wheel behaved WT-like with regards to food intake, energy expenditure, respiratory exchange ratio and locomotion regardless of being fed on a chow or high fat diet. Interestingly however, when given the access to a running wheel, Npy4r-/- mice while having a comparable locomotor activity, showed significantly higher wheel-running activity than WT, again regardless of dietary conditions. This higher wheel-running activity in Npy4r-/-mice arose from an increased dark-phase running time rather than changes in number of running bouts or the running speed. Consistently, energy expenditure was higher in Npy4r-/- than WT mice. Importantly, food intake was reduced in Npy4r-/-mice under wheel access condition which was due to decreased feeding bouts rather than changes in meal size. Together, these findings demonstrate an important role of Npy4r signalling in the dual control of feeding and physical activity, particularly in the form of wheel-running activity.


Subject(s)
Eating , Energy Metabolism , Feeding Behavior , Mice, Knockout , Neuropeptide Y , Receptors, Neuropeptide Y , Signal Transduction , Animals , Receptors, Neuropeptide Y/metabolism , Receptors, Neuropeptide Y/genetics , Signal Transduction/physiology , Neuropeptide Y/metabolism , Energy Metabolism/physiology , Feeding Behavior/physiology , Eating/physiology , Mice , Male , Mice, Inbred C57BL , Motor Activity/physiology , Physical Conditioning, Animal/physiology , Diet, High-Fat , Locomotion/physiology
15.
Neuropeptides ; 104: 102412, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38330680

ABSTRACT

Neuropeptide Y (NPY), an extensively distributed neurotransmitter within the central nervous system (CNS), was initially detected and isolated from the brain of a pig in 1982. By binding to its G protein-coupled receptors, NPY regulates immune responses and contributes to the pathogenesis of numerous inflammatory diseases. The hippocampus contained the maximum concentration in the CNS, with the cerebral cortex, hypothalamus, thalamus, brainstem, and cerebellum following suit. This arrangement suggests that the substance has a specific function within the CNS. More and more studies have shown that NPY is involved in the physiological and pathological mechanism of stroke, and its serum concentration can be one of the specific biomarkers of stroke and related complications because of its high activity, broad and complex effects. By summarizing relevant literature, this article aims to gain a thorough understanding of the potential clinical applications of NPY in the treatment of stroke, identification of stroke and its related complications, and assessment of prognosis.


Subject(s)
Neuropeptide Y , Receptors, Neuropeptide Y , Stroke , Animals , Neuropeptide Y/metabolism , Neuropeptide Y/therapeutic use , Prognosis , Receptors, Neuropeptide Y/metabolism , Signal Transduction , Stroke/diagnosis , Stroke/therapy , Swine , Humans
16.
ESC Heart Fail ; 11(3): 1625-1635, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38400690

ABSTRACT

AIMS: Cardiac dysfunction is commonly observed in patients with subarachnoid haemorrhage (SAH). However, the specific timeline of cardiac remodelling and the underlying mechanisms responsible for this effect following SAH remain unknown. This study aims to explore the impact of SAH on cardiac dysfunction and its potential mechanisms over time. METHODS AND RESULTS: In Protocol 1, we investigated cardiac function and potential mechanisms in a Sprague-Dawley rat model of SAH at six time points (baseline and Days 1, 3, 7, 14, and 28) while exploring the underlying mechanisms. Our assessments included the haemodynamic profile, echocardiography, and the concentrations of plasma biomarkers at various time points post-SAH. We determined neuropeptide Y (NPY) 1-5 receptor protein expression levels through western blotting. In Protocol 2, we administered an NPY1 receptor antagonist to evaluate the effects of cardiac dysfunction induced by SAH on Day 3. In Protocol 1, SAH gradually provoked cardiac systolic dysfunction during the acute phase, reaching its peak on Day 3 without concurrent alterations in wall thickness. However, no significant changes were observed from Days 14 to 28 compared with Day 0. The changes in cardiac dysfunction were consistent with myocardial injury, inflammatory biomarkers, and NPY levels. SAH resulted in a heightened heart rate and systolic blood pressure, correlating with elevated epinephrine and norepinephrine levels. In Protocol 2, the administration of the NPY1 receptor antagonist effectively ameliorated cardiac dysfunction. CONCLUSIONS: SAH induces transient cardiac dysfunction in the acute phase, and the underlying mechanisms for this response involve the NPY-NPY1 receptor pathway, otherwise known as catecholamines.


Subject(s)
Disease Models, Animal , Rats, Sprague-Dawley , Receptors, Neuropeptide Y , Subarachnoid Hemorrhage , Animals , Subarachnoid Hemorrhage/complications , Subarachnoid Hemorrhage/physiopathology , Rats , Male , Receptors, Neuropeptide Y/antagonists & inhibitors , Receptors, Neuropeptide Y/metabolism , Time Factors , Echocardiography , Biomarkers/blood , Neuropeptide Y/metabolism , Ventricular Remodeling/physiology
17.
Endocrinology ; 165(5)2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38368624

ABSTRACT

Glucoprivic feeding is one of several counterregulatory responses (CRRs) that facilitates restoration of euglycemia following acute glucose deficit (glucoprivation). Our previous work established that glucoprivic feeding requires ventrolateral medullary (VLM) catecholamine (CA) neurons that coexpress neuropeptide Y (NPY). However, the connections by which VLM CA/NPY neurons trigger increased feeding are uncertain. We have previously shown that glucoprivation, induced by an anti-glycolygic agent 2-deoxy-D-glucose (2DG), activates perifornical lateral hypothalamus (PeFLH) neurons and that expression of NPY in the VLM CA/NPY neurons is required for glucoprivic feeding. We therefore hypothesized that glucoprivic feeding and possibly other CRRs require NPY-sensitive PeFLH neurons. To test this, we used the ribosomal toxin conjugate NPY-saporin (NPY-SAP) to selectively lesion NPY receptor-expressing neurons in the PeFLH of male rats. We found that NPY-SAP destroyed a significant number of PeFLH neurons, including those expressing orexin, but not those expressing melanin-concentrating hormone. The PeFLH NPY-SAP lesions attenuated 2DG-induced feeding but did not affect 2DG-induced increase in locomotor activity, sympathoadrenal hyperglycemia, or corticosterone release. The 2DG-induced feeding response was also significantly attenuated in NPY-SAP-treated female rats. Interestingly, PeFLH NPY-SAP lesioned male rats had reduced body weights and decreased dark cycle feeding, but this effect was not seen in female rats. We conclude that a NPY projection to the PeFLH is necessary for glucoprivic feeding, but not locomotor activity, hyperglycemia, or corticosterone release, in both male and female rats.


Subject(s)
Feeding Behavior , Hypothalamus , Neurons , Neuropeptide Y , Rats, Sprague-Dawley , Animals , Female , Male , Rats , Deoxyglucose/pharmacology , Eating/drug effects , Eating/physiology , Feeding Behavior/drug effects , Glucose/metabolism , Hypothalamic Area, Lateral/metabolism , Hypothalamic Area, Lateral/drug effects , Hypothalamic Hormones/metabolism , Hypothalamus/metabolism , Hypothalamus/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Melanins/metabolism , Neurons/metabolism , Neurons/drug effects , Neuropeptide Y/metabolism , Neuropeptide Y/pharmacology , Neuropeptides/metabolism , Orexins/metabolism , Pituitary Hormones/metabolism , Receptors, Neuropeptide Y/metabolism , Receptors, Neuropeptide Y/genetics , Ribosome Inactivating Proteins, Type 1/pharmacology , Saporins/pharmacology
18.
J Phys Chem Lett ; 15(6): 1711-1718, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38319949

ABSTRACT

The structure and dynamics of the lipid membrane can affect the activity of membrane proteins. Therefore, small lipophilic molecules that alter membrane properties (such as the neurotransmitter serotonin) can potentially modulate receptor activity without binding to the receptor. Here, we investigated how the activity of neuropeptide Y type 4 receptor (Y4R, reconstituted in lipid bicelles) is modulated by serotonin, which has no known interaction with Y4R. We found a serotonin-concentration-dependent decrease (down to 0.1 mM of serotonin) in the ligand affinity of Y4R. This effect correlates with a serotonin-induced reduction of the resistance of the bilayer to indentation (measured by atomic force microscopy) and bilayer thickness (measured by solid state NMR) in two different types of zwitterionic lipid bicelles. Our findings indicate a "membrane-mediated allosteric effect" of serotonin on the activation of Y4R and suggest the potential for developing pharmacophores, which can modulate cellular signaling without directly interacting with any receptor.


Subject(s)
Receptors, G-Protein-Coupled , Serotonin , Receptors, Neuropeptide Y/metabolism , Membrane Proteins/chemistry , Lipids , Lipid Bilayers/chemistry
19.
Neuropharmacology ; 246: 109847, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38218578

ABSTRACT

Neuropeptide Y (NPY) has anxiolytic-like effects and facilitates the extinction of cued and contextual fear in rodents. We have previously shown that intracerebroventricular administration of NPY reduces the expression of social fear via simultaneous activation of Y1 and Y2 receptors in a mouse model of social fear conditioning (SFC). In the present study, we investigated whether the anteroventral bed nucleus of the stria terminalis (BNSTav) mediates these effects of NPY, given the important role of BNSTav in regulating anxiety- and fear-related behaviors. We show that while NPY (0.1 nmol/0.2 µl/side) did not reduce the expression of SFC-induced social fear in male CD1 mice, it reduced the expression of both cued and contextual fear by acting on Y2 but not on Y1 receptors within the BNSTav. Prior administration of the Y2 receptor antagonist BIIE0246 (0.2 nmol/0.2 µl/side) but not of the Y1 receptor antagonist BIBO3304 trifluoroacetate (0.2 nmol/0.2 µl/side) blocked the effects of NPY on the expression of cued and contextual fear. Similarly, NPY exerted non-social anxiolytic-like effects in the elevated plus maze test but not social anxiolytic-like effects in the social approach avoidance test by acting on Y2 receptors and not on Y1 receptors within the BNSTav. These results suggest that administration of NPY within the BNSTav exerts robust Y2 receptor-mediated fear-reducing and anxiolytic-like effects specifically in non-social contexts and add a novel piece of evidence regarding the neural underpinnings underlying the effects of NPY on conditioned fear and anxiety-like behavior.


Subject(s)
Anterior Thalamic Nuclei , Anti-Anxiety Agents , Septal Nuclei , Male , Mice , Animals , Neuropeptide Y/pharmacology , Neuropeptide Y/metabolism , Receptors, Neuropeptide Y/metabolism , Anti-Anxiety Agents/pharmacology , Septal Nuclei/metabolism , Anxiety/drug therapy , Fear , Anterior Thalamic Nuclei/metabolism
20.
Theranostics ; 14(1): 363-378, 2024.
Article in English | MEDLINE | ID: mdl-38164144

ABSTRACT

Rationale: In the physiological states, the act of scratching protects the person from harmful substances, while in certain pathological conditions, the patient suffers from chronic itch, both physically and mentally. Chronic itch sufferers are more sensitive to mechanical stimuli, and mechanical hyperknesis relief is essential for chronic itch treatment. While neuropeptide Y-Y1 receptor (NPY-Y1R) system is known to play a crucial role in modulating mechanical itch in physiological conditions, it is elusive how they are altered during chronic itch. We hypothesize that the negative regulatory effect of Y1Rs on Tac2 neurons, the key neurons that transmit mechanical itch, declines during chronic itch. Methods: We combined transgenic mice, chemogenetic manipulation, immunofluorescence, rabies virus circuit tracing, and electrophysiology to investigate the plasticity of Y1Rs on Tac2 neurons during chronic itch. Results: We found that Tac2 neurons receive direct input from Npy neurons and that inhibition of Npy neurons induces activation of Tac2 neurons. Moreover, the expression of Y1Rs on Tac2 neurons is reduced, and the regulatory effect is also reduced during chronic itch. Conclusion: Our study clarifies the plasticity of Y1Rs on Tac2 neurons during chronic itch and further elucidates the mechanism by which NPY-Y1R system is responsible for modulating mechanical itch. We highlight Y1Rs as a promising therapeutic target for mechanical hyperknesis during chronic itch.


Subject(s)
Neuropeptide Y , Receptors, Neuropeptide Y , Humans , Mice , Animals , Neuropeptide Y/metabolism , Neuropeptide Y/pharmacology , Receptors, Neuropeptide Y/genetics , Receptors, Neuropeptide Y/metabolism , Neurons/metabolism , Pruritus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...