Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 219
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Anesthesiology ; 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39121458

ABSTRACT

BACKGROUND: Neuropeptide Y (NPY) Y2 receptor (Y2) antagonist BIIE0246 can both inhibit and facilitate nociception. We hypothesized that Y2 function depends on inflammation or nerve injury status. METHODS: We implemented a battery of behavioral tests in mice of both sexes that received: 1) no injury; 2) an incision model of postoperative pain; 3) a spared nerve injury (SNI) model of neuropathic pain; and 4) a latent sensitization model of chronic postsurgical pain. In addition to Y2 gene expression assays, spinal Y2 G-protein coupling was studied with [ 35S]GTPγS binding assays. RESULTS: We report that intrathecal BIIE0246 increased mechanical and cold hypersensitivity, produced behavioral signs of spontaneous nociception and itch, and produced conditioned place aversion and preference in normal, uninjured mice. BIIE0246 did not change heat hypersensitivity or motor coordination. Conditional (sensory neuron-specific) Y2 deletion prevented BIIE0246-induced mechanical and cold hypersensitivity, nocifensive behaviors and aversion. Both conditional deletion and pharmacological blockade of Y2 reduced mechanical and thermal hypersensitivity after incision or nerve injury. SNI did not change the sensitivity of Y2 G-protein coupling with the Y2 agonist PYY3-36, but increased the population of Y2 that effectively coupled G-proteins. Intrathecal PYY3-36 failed to reduce SNI- or incision-induced hypersensitivity in C57BL/6N mice. Incision did not change Npy2r gene expression in dorsal root ganglion. CONCLUSIONS: We conclude that Y2 at central terminals of primary afferent neurons provide tonic inhibition of mechanical and cold nociception and itch. This switches to the promotion of mechanical and thermal hyperalgesia in models of acute and chronic postsurgical and neuropathic pain, perhaps due to an increase in the population of Y2 that effectively couple to G-proteins. These results support the development of Y2 antagonists for the treatment of chronic postsurgical and neuropathic pain.

2.
Neurochem Res ; 48(8): 2490-2501, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37017888

ABSTRACT

The nucleus accumbens shell is a critical node in reward circuitry, encoding environments associated with reward. Long-range inputs from the ventral hippocampus (ventral subiculum) to the nucleus accumbens shell have been identified, yet their precise molecular phenotype remains to be determined. Here we used retrograde tracing to identify the ventral subiculum as the brain region with the densest glutamatergic (VGluT1-Slc17a7) input to the shell. We then used circuit-directed translating ribosome affinity purification to examine the molecular characteristics of distinct glutamatergic (VGluT1, VGluT2-Slc17a6) ventral subiculum to nucleus accumbens shell projections. We immunoprecipitated translating ribosomes from this population of projection neurons and analysed molecular connectomic information using RNA sequencing. We found differential gene enrichment across both glutamatergic projection neuron subtypes. In VGluT1 projections, we found enrichment of Pfkl, a gene involved in glucose metabolism. In VGluT2 projections, we found a depletion of Sparcl1 and Dlg1, genes known to play a role in depression- and addiction-related behaviours. These findings highlight potential glutamatergic neuronal-projection-specific differences in ventral subiculum to nucleus accumbens shell projections. Together these data advance our understanding of the phenotype of a defined brain circuit.


Subject(s)
Hippocampus , Nucleus Accumbens , Brain , Hippocampus/metabolism , Nucleus Accumbens/metabolism , Reward , Animals , Mice
3.
Diabetologia ; 65(6): 984-996, 2022 06.
Article in English | MEDLINE | ID: mdl-35316840

ABSTRACT

AIMS/HYPOTHESIS: Pancreatic beta cell dedifferentiation, transdifferentiation into other islet cells and apoptosis have been implicated in beta cell failure in type 2 diabetes, although the mechanisms are poorly defined. The endoplasmic reticulum stress response factor X-box binding protein 1 (XBP1) is a major regulator of the unfolded protein response. XBP1 expression is reduced in islets of people with type 2 diabetes, but its role in adult differentiated beta cells is unclear. Here, we assessed the effects of Xbp1 deletion in adult beta cells and tested whether XBP1-mediated unfolded protein response makes a necessary contribution to beta cell compensation in insulin resistance states. METHODS: Mice with inducible beta cell-specific Xbp1 deletion were studied under normal (chow diet) or metabolic stress (high-fat diet or obesity) conditions. Glucose tolerance, insulin secretion, islet gene expression, alpha cell mass, beta cell mass and apoptosis were assessed. Lineage tracing was used to determine beta cell fate. RESULTS: Deletion of Xbp1 in adult mouse beta cells led to beta cell dedifferentiation, beta-to-alpha cell transdifferentiation and increased alpha cell mass. Cell lineage-specific analyses revealed that Xbp1 deletion deactivated beta cell identity genes (insulin, Pdx1, Nkx6.1, Beta2, Foxo1) and derepressed beta cell dedifferentiation (Aldh1a3) and alpha cell (glucagon, Arx, Irx2) genes. Xbp1 deletion in beta cells of obese ob/ob or high-fat diet-fed mice triggered diabetes and worsened glucose intolerance by disrupting insulin secretory capacity. Furthermore, Xbp1 deletion increased beta cell apoptosis under metabolic stress conditions by attenuating the antioxidant response. CONCLUSIONS/INTERPRETATION: These findings indicate that XBP1 maintains beta cell identity, represses beta-to-alpha cell transdifferentiation and is required for beta cell compensation and prevention of diabetes in insulin resistance states.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Insulin-Secreting Cells , X-Box Binding Protein 1/metabolism , Animals , Cell Transdifferentiation/genetics , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Humans , Insulin/metabolism , Insulin Resistance/genetics , Insulin-Secreting Cells/metabolism , Mice , Stress, Physiological , X-Box Binding Protein 1/genetics
4.
Int J Obes (Lond) ; 46(10): 1749-1758, 2022 10.
Article in English | MEDLINE | ID: mdl-35794191

ABSTRACT

BACKGROUND/OBJECTIVE: Female mice are often excluded from diet-induced obesity studies as they are more resistant to the obesifying effects of a high-fat diet (HFD). However, the underlying mechanisms behind this sex disparity may actually have important implications for the development and management of obesity in humans. Therefore, we systematically investigated the immediate sex-specific effects of transitioning to a HFD in C57BL/6J mice as well as monitored whether these effects are altered after sustained HFD feeding and whether sex affects the response to a return to chow, representative of dieting. METHODS: Dual X-ray absorptiometry (DXA) analysis of body composition, indirect calorimetry measurements, and qPCR analysis of hypothalamic and brainstem regions were performed on male and female C57BL/6J mice. RESULTS: HFD had immediate and dramatic effects in males, increasing fat mass by 58% in the first 3 days. The resistance to the obesifying effect of HFD in females was linked both to an ability to maintain activity levels as well as to an immediate and significantly enhanced reduction in respiratory quotient (RQ), suggesting a greater ability to utilise fat in the diet as a source of fuel. Mechanistically, this sex disparity may be at least partially due to inherent sex differences in the catabolic (POMC/CART) versus anabolic (NPY/AgRP) neurological signalling pathways. Interestingly, the reintroduction of chow following HFD had immediate and consistent responses between the sexes with body composition and most metabolic parameters normalised within 3 days. However, both sexes displayed elevated hypothalamic Npy levels reminiscent of starvation. The difference in RQ seen between the sexes on HFD was immediately abolished suggesting similar abilities to burn fat reserves for fuel. CONCLUSIONS: C57BL/6J mice have markedly different sex-specific behavioural and metabolic responses to the introduction as well as the sustained intake of a HFD, but consistent responses to a dieting situation.


Subject(s)
Diet, High-Fat , Dietary Fats , Obesity , Agouti-Related Protein/metabolism , Animals , Diet, High-Fat/adverse effects , Dietary Fats/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Obesity/etiology , Obesity/metabolism , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism , Sex Factors
5.
FASEB J ; 35(11): e21980, 2021 11.
Article in English | MEDLINE | ID: mdl-34694651

ABSTRACT

Although best known for their involvement in modulating nociception, Neuropeptide FF (NPFF) group peptides have been suggested to fulfil a variety of biological functions such as feeding, anxiety behaviors and thermogenesis. However, evidence supporting these functions of NPFF is mostly pharmacological, leaving the physiological relevance unaddressed. Here we examined the physiological impact of lack of NPFF signalling in both genders using a Npff-/- mouse model. NPFF expression in the mouse is restricted to the spinal cord and brainstem while its cognate receptor NPFFR2 has wider distribution throughout the brain. Both male and female Npff-/- mice showed reduced repetitive behaviors evidenced in the marble burying test and self-grooming test. A decrease in anxiety-related behaviors in the Npff-/- mice was also observe in the open field test and to a lesser degree in an elevated plus maze test. Moreover, both male and female Npff-/- mice exhibited increased water intake resulting from increases in drinking size, rather than number of drinking events. During a fasting-refeeding challenge, Npff-/- mice of both genders displayed alterations in reparatory exchange ratio that reflect a greater fuel type flexibility. Npff-/- mice were otherwise wild-type-like regarding body weight, body composition, feeding behaviors, locomotion or energy expenditure. Together, these findings reveal the important physiological roles of NPFF signalling in the regulation of anxiety-related and repetitive behaviors, fluid homeostasis and oxidative fuel selection, highlighting the therapeutical potential of the NPFF system in a number of behavioral and metabolic disorders.


Subject(s)
Anxiety/metabolism , Drinking Behavior , Oligopeptides/physiology , Receptors, Neuropeptide/metabolism , Animals , Body Weight , Energy Metabolism , Female , Male , Mice , Mice, Knockout
6.
FASEB J ; 35(4): e21435, 2021 04.
Article in English | MEDLINE | ID: mdl-33749879

ABSTRACT

Peptide YY (PYY), produced by endocrine L cells in the gut, is known for its critical role in regulating gastrointestinal functions as well as satiety. However, how these processes are integrated with maintaining a healthy gut microbiome composition is unknown. Here, we show that lack of PYY in mice leads to distinct changes in gut microbiome composition that are diet-dependent. While under chow diet only slight differences in gut microbiome composition could be observed, high-fat diet (HFD) aggravated these differences. Specifically an increased abundance of the Bacteroidetes phylum with a corresponding decrease of the Firmicutes/Bacteroidetes ratio could be detected in Pyy-knockout (KO) mice in response to HFD. Detailed analysis of the Bacteroidetes phylum further revealed that the Alistipes genus belonging to the Rikenellaceae family, the Parabacteroides belonging to the Tannerellaceae family, as well as Muribaculum were increased in Pyy-KO mice. In order to investigate whether these changes are associated with changed markers of gut barrier and immunity, we analyzed the colonic expression of various pro-inflammatory cytokines, as well as tight junction proteins and mucin 2, and identified increased mRNA expression of the tight junction proteins Cldn2 and Ocel1 in Pyy-KO mice, while pro-inflammatory cytokine expression was not significantly altered. Together these results highlight a critical gene-environment interaction between diet and the gut microbiome and its impact on homeostasis of the intestinal epithelium under conditions of reduced PYY signaling which is commonly seen under obese conditions.


Subject(s)
Bacteria/classification , Diet, High-Fat/adverse effects , Gastrointestinal Microbiome , Peptide YY/metabolism , Animals , Body Composition , Mice , Mice, Knockout , Peptide YY/genetics
7.
J Integr Neurosci ; 21(1): 6, 2022 Jan 28.
Article in English | MEDLINE | ID: mdl-35164442

ABSTRACT

Insulin is known to act in the central nervous system to regulate several physiological and behavioural outcomes, including energy balance, glucose homeostasis and cognitive functioning. However, the neuronal populations through which insulin enhances cognitive performance remain unidentified. Insulin receptors are found in neuropeptide-Y (NPY) expressing neurons, which are abundant in the hypothalamus and hippocampus; regions involved in feeding behaviour and spatial memory, respectively. Here we show that mice with a tissue specific knockout of insulin receptors in NPY expressing neurons (IRl⁢o⁢x/l⁢o⁢x; NPYC⁢r⁢e⁣/+) display an impaired performance in the probe trial of the Morris Water Maze compared with control mice at both the 6 and the 12, but not at the 24 months time point, consistent with a crucial role of insulin and NPY in cognitive functioning. By 24 months of age all groups demonstrated similar reductions in spatial memory performance. Together, these data suggest that the mechanisms through which insulin influences cognitive functioning are, at least in part, via insulin receptor signaling in NPY expressing neurons. These results also highlight that cognitive impairments observed in aging may be due to impaired insulin signaling.


Subject(s)
Aging/physiology , Cognitive Dysfunction , Hippocampus , Neurons/metabolism , Neuropeptide Y/metabolism , Receptor, Insulin/physiology , Aging/metabolism , Animals , Behavior, Animal/physiology , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/physiopathology , Disease Models, Animal , Hippocampus/metabolism , Hippocampus/physiopathology , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Insulin/deficiency , Spatial Memory/physiology
8.
FASEB J ; 34(3): 4163-4177, 2020 03.
Article in English | MEDLINE | ID: mdl-31960508

ABSTRACT

Genetic and pharmacological functional studies have provided evidence that the lack of Neuropeptide Y-Y1  receptor (Y1 R) signaling pathway induces a high bone mass phenotype in mice. However, clinical observations have shown that drug or genetic mediated improvement of bone mass might be associated to alterations to bone extracellular matrix (ECM) properties, leading to bone fragility. Hence, in this study we propose to characterize the physical, chemical and biomechanical properties of mature bone ECM of germline NPY-Y1 R knockout (Y1 R-/- ) mice, and compare to their wild-type (WT) littermates. Our results demonstrated that the high bone mass phenotype observed in Y1 R-/- mice involves alterations in Y1 R-/-  bone ECM ultrastructure, as a result of accelerated deposition of organic and mineral fractions. In addition, Y1 R-/- bone ECM displays enhanced matrix maturation characterized by greater number of mature/highly packed collagen fibers without pathological accumulation of immature/mature collagen crosslinks nor compromise of mineral crystallinity. These unique features of Y1 R-/-  bone ECM improved the biochemical properties of Y1 R-/-  bones, reflected by mechanically robust bones with diminished propensity to fracture, contributing to greater bone strength. These findings support the future usage of drugs targeting Y1 R signaling as a promising therapeutic strategy to treat bone loss-related pathologies.


Subject(s)
Bone Matrix/metabolism , Receptors, Neuropeptide Y/metabolism , Animals , Body Weight/genetics , Body Weight/physiology , Enzyme-Linked Immunosorbent Assay , Male , Mechanical Tests , Mice , Mice, Knockout , Microscopy, Confocal , Microscopy, Electron, Transmission , Receptors, Neuropeptide Y/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Spectrum Analysis, Raman , X-Ray Microtomography
9.
Horm Behav ; 136: 105056, 2021 11.
Article in English | MEDLINE | ID: mdl-34509673

ABSTRACT

Mammalian taste bud cells express receptors for numerous peptides implicated elsewhere in the body in the regulation of metabolism, nutrient assimilation, and satiety. The perturbation of several peptide signaling pathways in the gustatory periphery results in changes in behavioral and/or physiological responsiveness to subsets of taste stimuli. We previously showed that Peptide YY (PYY) - which is present in both saliva and in subsets of taste cells - can affect behavioral taste responsiveness and reduce food intake and body weight. Here, we investigated the contributions of taste bud-localized receptors for PYY and the related Neuropeptide Y (NPY) on behavioral taste responsiveness. Y1R, but not Y2R, null mice show reduced responsiveness to sweet, bitter, and salty taste stimuli in brief-access taste tests; similar results were seen when wildtype mice were exposed to Y receptor antagonists in the taste stimuli. Finally, mice in which the gene encoding the NPY propeptide was deleted also showed reduced taste responsiveness to sweet and bitter taste stimuli. Collectively, these results suggest that Y1R signaling, likely through its interactions with NPY, can modulate peripheral taste responsiveness in mice.


Subject(s)
Taste Buds , Taste , Animals , Male , Mammals/metabolism , Mice , Mice, Knockout , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Peptide YY/metabolism , Receptors, Neuropeptide Y/genetics , Receptors, Neuropeptide Y/metabolism , Taste Buds/metabolism
10.
Int J Obes (Lond) ; 44(10): 2149-2164, 2020 10.
Article in English | MEDLINE | ID: mdl-32152498

ABSTRACT

BACKGROUND/OBJECTIVES: Maintaining energy balance is important to ensure a healthy organism. However, energy partitioning, coordinating the distribution of sufficient energy to different organs and tissues is equally important, but the control of this process is largely unknown. In obesity, an increase in fat mass necessitates the production of additional bone mass to cope with the increase in bodyweight and processes need to be in place to communicate this new weight bearing demand. Here, we investigate the interaction between leptin and NPY, two factors critically involved in the regulation of both energy metabolism and bone mass, in this process. METHODS: We assessed the co-localization of leptin receptors on NPY neurons using RNAScope followed by a systematic examination of body composition and energy metabolism profiling in male and female mice lacking leptin receptors specifically in NPY neurons (Leprlox/lox;NPYCre/+). The effect of short-term switching between chow and high-fat diet was also examined in these mice. RESULTS: We uncovered that leptin receptor expression is greater on a subpopulation of NPY neurons in the arcuate that do not express AgRP. We further show that Leprlox/lox;NPYCre/+ mice exhibit significantly increased adiposity while bone mass is diminished. These body composition changes occur in the absence of alterations in food intake or energy expenditure, demonstrating a prominent role for leptin signaling in NPY neurons in the control of energy partitioning. Importantly however, when fed a high-fat diet, these mice display a switch in energy partitioning whereby they exhibit a significantly enhanced ability to increase their bone mass to match the increased bodyweight caused by higher caloric intake concurrent with attenuated adiposity. CONCLUSIONS: Taken together, these results demonstrate that leptin signaling in NPY neurons is critical for coordinating energy partitioning between fat and bone mass especially during situations of changes in energy balance.


Subject(s)
Adipose Tissue/metabolism , Bone and Bones/metabolism , Energy Metabolism , Hypothalamus/metabolism , Leptin/metabolism , Neurons/metabolism , Adiposity , Animals , Body Composition , Diet, High-Fat , Energy Intake , Female , Male , Mice , Receptors, Leptin
11.
Calcif Tissue Int ; 107(5): 510-523, 2020 11.
Article in English | MEDLINE | ID: mdl-32804252

ABSTRACT

The maintenance of whole body energy homeostasis is critical to survival and mechanisms exist whereby an organism can adapt to its environment and the stresses placed upon it. Environmental temperature and thermogenesis are key components known to affect energy balance. However, little is known about how these processes are balanced against the overall energy balance. We show that even mild cold exposure has a significant effect on energy expenditure and UCP-1 levels which increase by 43% and 400%, respectively, when wild-type (WT) mice at thermoneutral (29 °C) were compared to mice at room temperature (22 °C) conditions. Interestingly, bone mass was lower in cold-stressed WT mice with significant reductions in femoral bone mineral content (- 19%) and bone volume (- 13%). Importantly, these cold-induced skeletal changes were absent in mice lacking NPY, one of the main controllers of energy homeostasis, highlighting the critical role of NPY in this process. However, energy expenditure was significantly greater in cold-exposed NPY null mice, indicating that suppression of non-thermogenic tissues, like bone, contributes to the adaptive responses to cold exposure. Altogether, this work identifies NPY as being crucial in coordinating energy and bone homeostasis where it suppresses energy expenditure, UCP-1 levels and lowers bone mass under conditions of cold exposure.


Subject(s)
Bone Density , Cold Temperature , Energy Metabolism , Neuropeptide Y , Animals , Homeostasis , Mice , Neuropeptide Y/genetics , Uncoupling Protein 1
12.
EMBO J ; 34(12): 1648-60, 2015 Jun 12.
Article in English | MEDLINE | ID: mdl-25916827

ABSTRACT

Many reports have revealed the importance of the sympathetic nervous system (SNS) in the control of the bone marrow environment. However, the specific role of neuropeptide Y (NPY) in this process has not been systematically studied. Here we show that NPY-deficient mice have significantly reduced hematopoietic stem cell (HSC) numbers and impaired regeneration in bone marrow due to apoptotic destruction of SNS fibers and/or endothelial cells. Furthermore, pharmacological elevation of NPY prevented bone marrow impairments in a mouse model of chemotherapy-induced SNS injury, while NPY injection into conditional knockout mice lacking the Y1 receptor in macrophages did not relieve bone marrow dysfunction. These results indicate that NPY promotes neuroprotection and restores bone marrow dysfunction from chemotherapy-induced SNS injury through the Y1 receptor in macrophages. They also reveal a new role of NPY as a regulator of the bone marrow microenvironment and highlight the potential therapeutic value of this neuropeptide.


Subject(s)
Bone Marrow Cells/metabolism , Bone Marrow/physiology , Cellular Microenvironment/physiology , Hematopoietic Stem Cells/physiology , Nerve Fibers, Myelinated/metabolism , Neuropeptide Y/metabolism , Analysis of Variance , Animals , Apoptosis/physiology , Endothelial Cells/physiology , Flow Cytometry , Fluorescent Antibody Technique , Hematopoietic Stem Cells/metabolism , Mice , Mice, Knockout , Neuropeptide Y/deficiency , Sympathetic Nervous System/cytology
13.
Int J Obes (Lond) ; 42(11): 1925-1938, 2018 11.
Article in English | MEDLINE | ID: mdl-29523877

ABSTRACT

OBJECTIVE: To determine whether age and neuropeptide Y (NPY) were involved in the skeletal response to extended periods of diet-induced obesity. METHODS: Male wild-type (WT) and NPY null (NPYKO) mice were fed a mild (23% fat) high-fat diet for 10 weeks from 6 or 16 weeks of age. Metabolism and bone density were assessed during feeding. Skeletal changes were assessed by microCT and histomorphometry. RESULTS: High-fat feeding in 6-week-old WT mice led to significantly increased body weight, adiposity and serum leptin levels, accompanied with markedly suppressed cortical bone accrual. NPYKO mice were less susceptible to fat accrual but, importantly, displayed a complete lack of suppression of bone accrual or cortical bone loss. In contrast, when skeletally mature (16 week old) mice underwent 10 weeks of fat feeding, the metabolic response to HFD was similar to younger mice, however bone mass was not affected in either WT or NPYKO. Thus, growing mice are particularly susceptible to the detrimental effects of HFD on bone mass, through suppression of bone accrual involving NPY signalling. CONCLUSION: This study provides new insights into the relationship between the opposing processes of a positive weight/bone relationship and the negative 'metabolic' effect of obesity on bone mass. This negative effect is particularly active in growing skeletons, which have heightened sensitivity to changes in obesity. In addition, NPY is identified as a fundamental driver of this negative 'metabolic' pathway to bone.


Subject(s)
Bone Remodeling/physiology , Cortical Bone/pathology , Neuropeptide Y/deficiency , Obesity/pathology , Weight Gain/physiology , Animals , Bone Density , Diet, High-Fat/adverse effects , Disease Models, Animal , Mice , Mice, Inbred Strains , Neuropeptide Y/physiology , Obesity/metabolism
14.
Stem Cells ; 34(8): 2145-56, 2016 08.
Article in English | MEDLINE | ID: mdl-27090492

ABSTRACT

Hematopoietic stem/progenitor cell (HSPC) mobilization is an essential homeostatic process regulated by the interaction of cellular and molecular components in bone marrow niches. It has been shown by others that neurotransmitters released from the sympathetic nervous system regulate HSPC egress from bone marrow to peripheral blood. In this study, we investigate the functional role of neuropeptide Y (NPY) on this process. NPY deficient mice had significantly impaired HSPC mobilization due to increased expression of HSPC maintenance factors by reduction of matrix metalloproteinase-9 (MMP-9) activity in bone marrow. Pharmacological or endogenous elevation of NPY led to decrease of HSPC maintenance factors expression by activating MMP-9 in osteoblasts, resulting in HSPC mobilization. Mice in which the Y1 receptor was deleted in osteoblasts did not exhibit HSPC mobilization by NPY. Furthermore, NPY treatment in ovariectomized mice caused reduction of bone loss due to HSPC mobilization. These results suggest a new role of NPY on HSPC mobilization, as well as the potential therapeutic application of this neuropeptide for stem cell-based therapy. Stem Cells 2016;34:2145-2156.


Subject(s)
Hematopoietic Stem Cell Mobilization , Matrix Metalloproteinase 9/metabolism , Neuropeptide Y/metabolism , Osteoblasts/metabolism , Receptors, Neuropeptide Y/metabolism , Animals , Bone and Bones/metabolism , Chemotaxis , Female , Homeostasis , Mice, Inbred C57BL , Neuropeptide Y/deficiency , Osteoblasts/cytology , Osteoblasts/enzymology , Receptors, CXCR4/metabolism
15.
Bioconjug Chem ; 27(9): 2166-75, 2016 09 21.
Article in English | MEDLINE | ID: mdl-27513006

ABSTRACT

Traceable truncated Neuropeptide Y (NPY) analogues with Y1 receptor (Y1R) affinity and selectivity are highly desirable tools in studying receptor location, regulation, and biological functions. A range of fluorescently labeled analogues of a reported Y1R/Y4R preferring ligand BVD-15 have been prepared and evaluated using high content imaging techniques. One peptide, [Lys(2)(sCy5), Arg(4)]BVD-15, was characterized as an Y1R antagonist with a pKD of 7.2 measured by saturation analysis using fluorescent imaging. The peptide showed 8-fold lower affinity for Y4R (pKD = 6.2) and was a partial agonist at this receptor. The suitability of [Lys(2)(sCy5), Arg(4)]BVD-15 for Y1R and Y4R competition binding experiments was also demonstrated in intact cells. The nature of the label was shown to be critical with replacement of sCy5 by the more hydrophobic Cy5.5 resulting in a switch from Y1R antagonist to Y1R partial agonist.


Subject(s)
Carbocyanines/chemistry , Coloring Agents/chemistry , Neuropeptide Y/chemistry , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Receptors, Neuropeptide Y/metabolism , Amino Acid Sequence , Binding, Competitive , HEK293 Cells , Humans , Ligands , Staining and Labeling
16.
J Neurosci ; 34(49): 16309-19, 2014 Dec 03.
Article in English | MEDLINE | ID: mdl-25471570

ABSTRACT

The hypothalamic NPY system plays an important role in regulating food intake and energy expenditure. Different biological actions of NPY are assigned to NPY receptor subtypes. Recent studies demonstrated a close relationship between food intake and growth hormone (GH) secretion; however, the mechanism through which endogenous NPY modulates GH release remains unknown. Moreover, conclusive evidence demonstrating a role for NPY and Y-receptors in regulating the endogenous pulsatile release of GH does not exist. We used genetically modified mice (germline Npy, Y1, and Y2 receptor knock-out mice) to assess pulsatile GH secretion under both fed and fasting conditions. Deletion of NPY did not impact fed GH release; however, it reversed the fasting-induced suppression of pulsatile GH secretion. The recovery of GH secretion was associated with a reduction in hypothalamic somatotropin release inhibiting factor (Srif; somatostatin) mRNA expression. Moreover, observations revealed a differential role for Y1 and Y2 receptors, wherein the postsynaptic Y1 receptor suppresses GH secretion in fasting. In contrast, the presynaptic Y2 receptor maintains normal GH output under long-term ad libitum-fed conditions. These data demonstrate an integrated neural circuit that modulates GH release relative to food intake, and provide essential information to address the differential roles of Y1 and Y2 receptors in regulating the release of GH under fed and fasting states.


Subject(s)
Fasting/physiology , Growth Hormone/metabolism , Neuropeptide Y/physiology , Receptors, Neuropeptide Y/physiology , Animals , Blood Glucose , Growth Hormone-Releasing Hormone/biosynthesis , Hypothalamus/metabolism , Male , Mice , Mice, Knockout , Neuropeptide Y/blood , Neuropeptide Y/genetics , Peptide YY/blood , Receptors, Neuropeptide Y/genetics , Somatostatin/biosynthesis
17.
J Neurosci ; 33(31): 12792-804, 2013 Jul 31.
Article in English | MEDLINE | ID: mdl-23904614

ABSTRACT

Expression of dynorphin, an endogenous opioid peptide, increases with age and has been associated with memory impairments in rats. In human, prodynorphin (Pdyn) gene polymorphisms might be linked to cognitive function in the elderly. Moreover, elevated dynorphin levels have been reported in postmortem samples from Alzheimer's disease patients. However, the cellular and molecular processes affected by higher dynorphin levels during aging remain unknown. Using Pdyn(-/-) mice, we observed significant changes in the function and expression of Group 1 metabotropic glutamate receptor (mGluR). Compared with age-matched wild-type (WT) littermates, we found increased expression of mGluR1α and mGluR5 in the hippocampus and cortex of old, but not young, Pdyn(-/-) mice. Increased Group 1 mGluR expression in aged Pdyn(-/-) mice was associated with enhanced mGluR-mediated long-term depression, a form of synaptic plasticity. Notably, whereas aged WT mice developed spatial and recognition memory deficits, aged Pdyn(-/-) mice performed similarly as young mice. Pharmacological treatments with 3-cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide, a positive modulator of mGlu5 receptors, or norbinaltorphimine, an antagonist for dynorphin-targeted κ-opioid receptor, rescued memory in old WT mice. Conversely, mGlu5 receptor antagonist 2-methyl-6-(phenylethynyl)pyridine hydrochloride impaired spatial memory of old Pdyn(-/-) mice. Intact cognition in aged Pdyn(-/-) mice paralleled with increased expression of Group 1 mGluR-related genes Homer 1a and Arc. Finally, aged Pdyn(-/-) mice displayed less anxiety-related behaviors than age-matched WT mice. Together, our results suggest that elevated Pdyn expression during normal aging reduces mGluR expression and signaling, which in turn impairs cognitive functions and increases anxiety.


Subject(s)
Aging/physiology , Anxiety/metabolism , Enkephalins/deficiency , Gene Expression Regulation/genetics , Protein Precursors/deficiency , Receptors, Metabotropic Glutamate/metabolism , Animals , Anxiety/drug therapy , Benzamides/pharmacology , Benzamides/therapeutic use , Benzphetamine/analogs & derivatives , Benzphetamine/pharmacology , Central Nervous System Stimulants/pharmacology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cognition Disorders/drug therapy , Disease Models, Animal , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Amino Acid Antagonists/therapeutic use , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Gene Expression Regulation/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , In Vitro Techniques , Long-Term Synaptic Depression/drug effects , Long-Term Synaptic Depression/genetics , Memory Disorders/drug therapy , Memory Disorders/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Pyridines/pharmacology , Pyridines/therapeutic use , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Recognition, Psychology/drug effects , Recognition, Psychology/physiology
18.
J Neurosci ; 33(47): 18368-80, 2013 Nov 20.
Article in English | MEDLINE | ID: mdl-24259562

ABSTRACT

Hormone peptide tyrosine-tyrosine (PYY) is secreted into circulation from the gut L-endocrine cells in response to food intake, thus inducing satiation during interaction with its preferred receptor, Y2R. Clinical applications of systemically administered PYY for the purpose of reducing body weight were compromised as a result of the common side effect of visceral sickness. We describe here a novel approach of elevating PYY in saliva in mice, which, although reliably inducing strong anorexic responses, does not cause aversive reactions. The augmentation of salivary PYY activated forebrain areas known to mediate feeding, hunger, and satiation while minimally affecting brainstem chemoreceptor zones triggering nausea. By comparing neuronal pathways activated by systemic versus salivary PYY, we identified a metabolic circuit associated with Y2R-positive cells in the oral cavity and extending through brainstem nuclei into hypothalamic satiety centers. The discovery of this alternative circuit that regulates ingestive behavior without inducing taste aversion may open the possibility of a therapeutic application of PYY for the treatment of obesity via direct oral application.


Subject(s)
Feeding Behavior/drug effects , Peptide Fragments/pharmacology , Peptide YY/deficiency , Saliva/enzymology , Aminophylline , Animals , Conditioning, Psychological/drug effects , Eating/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Glucagon-Like Peptide 1/metabolism , Humans , Iodine Isotopes/pharmacokinetics , Male , Mice , Mice, Inbred C57BL , Oxytocin/metabolism , Peptide YY/chemistry , Proto-Oncogene Proteins c-fos/metabolism , Satiation/drug effects , Tyrosine 3-Monooxygenase/metabolism , Vasopressins/metabolism , alpha-MSH/metabolism
19.
Front Neuroendocrinol ; 34(2): 107-19, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23391702

ABSTRACT

Prader-Willi Syndrome (PWS) is a neurodevelopmental genetic disorder caused by loss of expression of imprinted, paternally inherited genes on chromosome 15q11q13. This imprinted gene cluster has its homologous region on mouse chromosome 7C. The extremely well conserved synteny between the human and the murine regions gave origin to the generation of mouse models for PWS, which facilitated investigations of the role and function of single genes or gene clusters in the pathogenesis of this disease. In this review we will describe which mouse models have been generated so far and how they were developed; we will focus on the consequences of single genes' (or gene clusters') loss of expression on the phenotype, highlighting the similarities to the human PWS features. PWS mouse models have brought major improvements in our knowledge about this complex condition, although the mechanisms implicated in its pathogenesis still remain not fully understood.


Subject(s)
Prader-Willi Syndrome/genetics , Animals , Antigens, Neoplasm/genetics , Chromosome Mapping , Chromosomes, Human, Pair 15 , Disease Models, Animal , Genomic Imprinting , Humans , Mice , Mice, Knockout , Multigene Family/genetics , Nerve Tissue Proteins/deficiency , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Proteins/genetics , RNA, Small Nucleolar/genetics , Synteny , snRNP Core Proteins/genetics
20.
FASEB J ; 27(12): 5022-33, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24043261

ABSTRACT

It has been hypothesized that the peripheral taste system may be modulated in the context of an animal's metabolic state. One purported mechanism for this phenomenon is that circulating gastrointestinal peptides modulate the functioning of the peripheral gustatory system. Recent evidence suggests endocrine signaling in the oral cavity can influence food intake (FI) and satiety. We hypothesized that these hormones may be affecting FI by influencing taste perception. We used immunohistochemistry along with genetic knockout models and the specific reconstitution of peptide YY (PYY) in saliva using gene therapy protocols to identify a role for PYY signaling in taste. We show that PYY is expressed in subsets of taste cells in murine taste buds. We also show, using brief-access testing with PYY knockouts, that PYY signaling modulates responsiveness to bitter-tasting stimuli, as well as to lipid emulsions. We show that salivary PYY augmentation, via viral vector therapy, rescues behavioral responsiveness to a lipid emulsion but not to bitter stimuli and that this response is likely mediated via activation of Y2 receptors localized apically in taste cells. Our findings suggest distinct functions for PYY produced locally in taste cells vs. that circulating systemically.


Subject(s)
Peptide YY/metabolism , Satiety Response , Taste , Animals , Eating , Mice , Mice, Knockout , Peptide YY/genetics , Receptors, Neuropeptide Y/genetics , Receptors, Neuropeptide Y/metabolism , Taste Buds/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL