Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
Aging (Albany NY) ; 8(9): 2153-2181, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27689748

ABSTRACT

We describe age-related molecular and neuronal changes that disrupt mobility or energy balance based on brain region and genetic background. Compared to young mice, aged C57BL/6 mice exhibit marked locomotor (but not energy balance) impairments. In contrast, aged BALB mice exhibit marked energy balance (but not locomotor) impairments. Age-related changes in cerebellar or hypothalamic gene expression accompany these phenotypes. Aging evokes upregulation of immune pattern recognition receptors and cell adhesion molecules. However, these changes do not localize to microglia, the major CNS immunocyte. Consistent with a neuronal role, there is a marked age-related increase in excitatory synapses over the cerebellum and hypothalamus. Functional imaging of these regions is consistent with age-related synaptic impairments. These studies suggest that aging reactivates a developmental program employed during embryogenesis where immune molecules guide synapse formation and pruning. Renewed activity in this program may disrupt excitatory neurotransmission, causing significant behavioral deficits.


Subject(s)
Aging/physiology , Cerebellum/physiology , Excitatory Amino Acids/physiology , Hypothalamus/physiology , Synapses/physiology , Synaptic Transmission/physiology , Aging/genetics , Aging/immunology , Animals , Energy Metabolism/physiology , Gene Expression , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Motor Activity/physiology
2.
Neuropsychopharmacology ; 41(5): 1404-15, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26383016

ABSTRACT

Elucidating how the brain's serotonergic network mediates diverse behavioral actions over both relatively short (minutes-hours) and long period of time (days-weeks) remains a major challenge for neuroscience. Our relative ignorance is largely due to the lack of technologies with robustness, reversibility, and spatio-temporal control. Recently, we have demonstrated that our chemogenetic approach (eg, Designer Receptors Exclusively Activated by Designer Drugs (DREADDs)) provides a reliable and robust tool for controlling genetically defined neural populations. Here we show how short- and long-term activation of dorsal raphe nucleus (DRN) serotonergic neurons induces robust behavioral responses. We found that both short- and long-term activation of DRN serotonergic neurons induce antidepressant-like behavioral responses. However, only short-term activation induces anxiogenic-like behaviors. In parallel, these behavioral phenotypes were associated with a metabolic map of whole brain network activity via a recently developed non-invasive imaging technology DREAMM (DREADD Associated Metabolic Mapping). Our findings reveal a previously unappreciated brain network elicited by selective activation of DRN serotonin neurons and illuminate potential therapeutic and adverse effects of drugs targeting DRN neurons.


Subject(s)
Anxiety/physiopathology , Depression/physiopathology , Dorsal Raphe Nucleus/physiology , Serotonergic Neurons/physiology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Brain/drug effects , Brain/metabolism , Brain/physiology , Circadian Rhythm , Designer Drugs/administration & dosage , Dorsal Raphe Nucleus/drug effects , Dorsal Raphe Nucleus/metabolism , Male , Mice , Mice, Transgenic , Serotonergic Neurons/drug effects , Serotonergic Neurons/metabolism , Time Factors
3.
PLoS One ; 10(12): e0142906, 2015.
Article in English | MEDLINE | ID: mdl-26630489

ABSTRACT

Although central serotonergic systems are known to influence responses to noxious stimuli, mechanisms underlying serotonergic modulation of pain responses are unclear. We proposed that serotonin 2C receptors (5-HT2CRs), which are expressed within brain regions implicated in sensory and affective responses to pain, contribute to the serotonergic modulation of pain responses. In mice constitutively lacking 5-HT2CRs (2CKO mice) we found normal baseline sensory responses to noxious thermal, mechanical and chemical stimuli. In contrast, 2CKO mice exhibited a selective enhancement of affect-related ultrasonic afterdischarge vocalizations in response to footshock. Enhanced affect-related responses to noxious stimuli were also exhibited by 2CKO mice in a fear-sensitized startle assay. The extent to which a brief series of unconditioned footshocks produced enhancement of acoustic startle responses was markedly increased in 2CKO mice. As mesolimbic dopamine pathways influence affective responses to noxious stimuli, and these pathways are disinhibited in 2CKO mice, we examined the sensitivity of footshock-induced enhancement of startle to dopamine receptor blockade. Systemic administration of the dopamine D2/D3 receptor antagonist raclopride selectively reduced footshock-induced enhancement of startle without influencing baseline acoustic startle responses. We propose that 5-HT2CRs regulate affective behavioral responses to unconditioned aversive stimuli through mechanisms involving the disinhibition of ascending dopaminergic pathways.


Subject(s)
Fear/physiology , Receptor, Serotonin, 5-HT2C/physiology , Reflex, Startle/physiology , Vocalization, Animal/physiology , Animals , Dopamine Antagonists/pharmacology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Raclopride/pharmacology , Receptors, Dopamine D2/chemistry , Reflex, Startle/drug effects , Ultrasonics , Vocalization, Animal/drug effects , Vocalization, Animal/radiation effects
4.
Proc Natl Acad Sci U S A ; 112(9): 2888-93, 2015 Mar 03.
Article in English | MEDLINE | ID: mdl-25695968

ABSTRACT

Gi-GPCRs, G protein-coupled receptors that signal via Gα proteins of the i/o class (Gαi/o), acutely regulate cellular behaviors widely in mammalian tissues, but their impact on the development and growth of these tissues is less clear. For example, Gi-GPCRs acutely regulate insulin release from pancreatic ß cells, and variants in genes encoding several Gi-GPCRs--including the α-2a adrenergic receptor, ADRA2A--increase the risk of type 2 diabetes mellitus. However, type 2 diabetes also is associated with reduced total ß-cell mass, and the role of Gi-GPCRs in establishing ß-cell mass is unknown. Therefore, we asked whether Gi-GPCR signaling regulates ß-cell mass. Here we show that Gi-GPCRs limit the proliferation of the insulin-producing pancreatic ß cells and especially their expansion during the critical perinatal period. Increased Gi-GPCR activity in perinatal ß cells decreased ß-cell proliferation, reduced adult ß-cell mass, and impaired glucose homeostasis. In contrast, Gi-GPCR inhibition enhanced perinatal ß-cell proliferation, increased adult ß-cell mass, and improved glucose homeostasis. Transcriptome analysis detected the expression of multiple Gi-GPCRs in developing and adult ß cells, and gene-deletion experiments identified ADRA2A as a key Gi-GPCR regulator of ß-cell replication. These studies link Gi-GPCR signaling to ß-cell mass and diabetes risk and identify it as a potential target for therapies to protect and increase ß-cell mass in patients with diabetes.


Subject(s)
Cell Proliferation , Diabetes Mellitus, Type 2/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Insulin-Secreting Cells/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Signal Transduction , Animals , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Glucose/genetics , Glucose/metabolism , Insulin-Secreting Cells/pathology , Mice , Mice, Transgenic , Receptors, Adrenergic, alpha-2/genetics
5.
Cell Stem Cell ; 14(4): 500-11, 2014 Apr 03.
Article in English | MEDLINE | ID: mdl-24561083

ABSTRACT

The ventricular-subventricular zone (V-SVZ) is an extensive germinal niche containing neural stem cells (NSCs) in the walls of the lateral ventricles of the adult brain. How the adult brain's neural activity influences the behavior of adult NSCs remains largely unknown. We show that serotonergic (5HT) axons originating from a small group of neurons in the raphe form an extensive plexus on most of the ventricular walls. Electron microscopy revealed intimate contacts between 5HT axons and NSCs (B1) or ependymal cells (E1) and these cells were labeled by a transsynaptic viral tracer injected into the raphe. B1 cells express the 5HT receptors 2C and 5A. Electrophysiology showed that activation of these receptors in B1 cells induced small inward currents. Intraventricular infusion of 5HT2C agonist or antagonist increased or decreased V-SVZ proliferation, respectively. These results indicate that supraependymal 5HT axons directly interact with NSCs to regulate neurogenesis via 5HT2C.


Subject(s)
Axons/physiology , Cell Differentiation , Neural Stem Cells/cytology , Neurons/physiology , Raphe Nuclei/physiology , Receptor, Serotonin, 5-HT2C/metabolism , Stem Cell Niche , Animals , Blotting, Western , Brain/cytology , Brain/physiology , Cell Proliferation , Electrophysiology , Immunoenzyme Techniques , Mice , Microscopy, Electron , Neural Stem Cells/metabolism , Neurogenesis , Neurons/cytology , RNA, Messenger/genetics , Raphe Nuclei/cytology , Real-Time Polymerase Chain Reaction , Receptor, Serotonin, 5-HT2C/chemistry , Reverse Transcriptase Polymerase Chain Reaction , Serotonin Receptor Agonists/pharmacology
6.
Front Neurosci ; 7: 36, 2013.
Article in English | MEDLINE | ID: mdl-23543912

ABSTRACT

Maintenance of energy balance requires regulation of the amount and timing of food intake. Decades of experiments utilizing pharmacological and later genetic manipulations have demonstrated the importance of serotonin signaling in this regulation. Much progress has been made in recent years in understanding how central nervous system (CNS) serotonin systems acting through a diverse array of serotonin receptors impact feeding behavior and metabolism. Particular attention has been paid to mechanisms through which serotonin impacts energy balance pathways within the hypothalamus. How upstream factors relevant to energy balance regulate the release of hypothalamic serotonin is less clear, but work addressing this issue is underway. Generally, investigation into the central serotonergic regulation of energy balance has had a predominantly "hypothalamocentric" focus, yet non-hypothalamic structures that have been implicated in energy balance regulation also receive serotonergic innervation and express multiple subtypes of serotonin receptors. Moreover, there is a growing appreciation of the diverse mechanisms through which peripheral serotonin impacts energy balance regulation. Clearly, the serotonergic regulation of energy balance is a field characterized by both rapid advances and by an extensive and diverse set of central and peripheral mechanisms yet to be delineated.

7.
Neuropsychopharmacology ; 38(6): 957-67, 2013 May.
Article in English | MEDLINE | ID: mdl-23303047

ABSTRACT

Although the serotonin (5-hydroxytryptamine, 5-HT) neurotransmitter system has been implicated in modulating executive control processes such as attention, response inhibition, and behavioral flexibility, the contributions of particular serotonin receptors remain unclear. Here, using operant-based behavioral paradigms, we demonstrate that mice with genetically ablated 5-HT2C receptors (2CKO mice) display deficits in executive functions. 2CKO mice were impaired in the acquisition of a visuospatial attention task as assessed in the 5-choice serial reaction time task (5-CSRTT). In this task, 2CKO mice exhibited marked impairment of attentional processes, with normal response inhibition. We assessed dynamic changes in neurotransmitter levels within the nucleus accumbens (NAc) by in vivo microdialysis in task-performing animals. Extracellular dopamine concentrations were elevated in the NAc of 2CKO mice during task performance, indicating that 5-HT2C receptors impact dopamine homeostasis during a visuospatial attention task. These findings raise the possibility that disinhibition of mesolimbic dopamine pathways contributes to impaired attention and perturbed task performance in 2CKO mice. Additionally, in a spatial reversal learning task, 2CKO mice failed to improve their performance over a series of reversals, indicating that intact 5-HT2C receptor signaling is required to accurately respond to repeated changes in reward contingencies. In contrast to the 2CKO phenotype in the 5-CSRTT, wild-type mice treated with the 5-HT2C receptor antagonist SB242084 exhibited diminished response inhibition, suggesting differing effects of acute pharmacological blockade and constitutive loss of 5-HT2C receptor activity. Altogether, these findings provide insights into the serotonergic regulation of executive control processes and suggest that impaired 5-HT2C receptor signaling during development may predispose to executive function disorders.


Subject(s)
Conditioning, Operant/physiology , Executive Function/physiology , Psychomotor Performance/physiology , Receptor, Serotonin, 5-HT2C/physiology , Animals , Conditioning, Operant/drug effects , Executive Function/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Psychomotor Performance/drug effects , Receptor, Serotonin, 5-HT2C/deficiency , Serotonin 5-HT2 Receptor Antagonists/pharmacology , Signal Transduction/genetics
8.
Diabetes ; 60(12): 3208-16, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22013016

ABSTRACT

OBJECTIVE: Despite their origins in different germ layers, pancreatic islet cells share many common developmental features with neurons, especially serotonin-producing neurons in the hindbrain. Therefore, we tested whether these developmental parallels have functional consequences. RESEARCH DESIGN AND METHODS: We used transcriptional profiling, immunohistochemistry, DNA-binding analyses, and mouse genetic models to assess the expression and function of key serotonergic genes in the pancreas. RESULTS: We found that islet cells expressed the genes encoding all of the products necessary for synthesizing, packaging, and secreting serotonin, including both isoforms of the serotonin synthetic enzyme tryptophan hydroxylase and the archetypal serotonergic transcription factor Pet1. As in serotonergic neurons, Pet1 expression in islets required homeodomain transcription factor Nkx2.2 but not Nkx6.1. In ß-cells, Pet1 bound to the serotonergic genes but also to a conserved insulin gene regulatory element. Mice lacking Pet1 displayed reduced insulin production and secretion and impaired glucose tolerance. CONCLUSIONS: These studies demonstrate that a common transcriptional cascade drives the differentiation of ß-cells and serotonergic neurons and imparts the shared ability to produce serotonin. The interrelated biology of these two cell types has important implications for the pathology and treatment of diabetes.


Subject(s)
Insulin-Secreting Cells/metabolism , Insulin/metabolism , Serotonin/metabolism , Animals , Cell Line , Cell Line, Tumor , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunohistochemistry , In Situ Hybridization , Insulin/genetics , Mice , NIH 3T3 Cells , Reverse Transcriptase Polymerase Chain Reaction , Serotonergic Neurons/metabolism , Serotonin/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Tryptophan Hydroxylase/genetics , Tryptophan Hydroxylase/metabolism , Zebrafish Proteins
9.
Genetics ; 189(1): 305-16, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21705752

ABSTRACT

In genetic studies, many interesting traits, including growth curves and skeletal shape, have temporal or spatial structure. They are better treated as curves or function-valued traits. Identification of genetic loci contributing to such traits is facilitated by specialized methods that explicitly address the function-valued nature of the data. Current methods for mapping function-valued traits are mostly likelihood-based, requiring specification of the distribution and error structure. However, such specification is difficult or impractical in many scenarios. We propose a general functional regression approach based on estimating equations that is robust to misspecification of the covariance structure. Estimation is based on a two-step least-squares algorithm, which is fast and applicable even when the number of time points exceeds the number of samples. It is also flexible due to a general linear functional model; changing the number of covariates does not necessitate a new set of formulas and programs. In addition, many meaningful extensions are straightforward. For example, we can accommodate incomplete genotype data, and the algorithm can be trivially parallelized. The framework is an attractive alternative to likelihood-based methods when the covariance structure of the data is not known. It provides a good compromise between model simplicity, statistical efficiency, and computational speed. We illustrate our method and its advantages using circadian mouse behavioral data.


Subject(s)
Chromosome Mapping , Phenotype , Quantitative Trait Loci , Algorithms , Animals , Behavior, Animal , Computer Simulation , Female , Genotype , Likelihood Functions , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Models, Genetic
10.
Pain ; 152(5): 990-1000, 2011 May.
Article in English | MEDLINE | ID: mdl-21256675

ABSTRACT

Despite the impact of chronic pain on the quality of life in patients, including changes to affective state and daily life activities, rodent preclinical models rarely address this aspect of chronic pain. To better understand the behavioral consequences of the tissue and nerve injuries typically used to model neuropathic and inflammatory pain in mice, we measured home cage and affective state behaviors in animals with spared nerve injury, chronic constriction injury (CCI), or intraplantar complete Freund's adjuvant. Mechanical hypersensitivity is prominent in each of these conditions and persists for many weeks. Home cage behavior was continuously monitored for 16 days in a system that measures locomotion, feeding, and drinking, and allows for precise analysis of circadian patterns. When monitored after injury, animals with spared nerve injury and complete Freund's adjuvant behaved no differently from controls in any aspect of daily life. Animals with CCI were initially less active, but the difference between CCI and controls disappeared by 2 weeks after injury. Further, in all pain models, there was no change in any measure of affective state. We conclude that in these standard models of persistent pain, despite the development of prolonged hypersensitivity, the mice do not have significantly altered "quality of life." As alteration in daily life activities is the feature that is so disrupted in patients with chronic pain, our results suggest that the models used here do not fully reflect the human conditions and point to a need for development of a murine chronic pain model in which lifestyle changes are manifest.


Subject(s)
Hyperalgesia/etiology , Inflammation/complications , Locomotion/physiology , Motor Activity/physiology , Sciatica/complications , Activities of Daily Living , Animals , Anxiety/etiology , Circadian Rhythm/physiology , Disease Models, Animal , Exploratory Behavior/physiology , Inflammation/chemically induced , Lipopolysaccharides/adverse effects , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mood Disorders/etiology , Pain Measurement
11.
J Biomed Sci ; 17: 82, 2010 Oct 17.
Article in English | MEDLINE | ID: mdl-20950489

ABSTRACT

BACKGROUND: Multicellular organisms are characterized by a remarkable diversity of morphologically distinct and functionally specialized cell types. Transgenic techniques for the manipulation of gene expression in specific cellular populations are highly useful for elucidating the development and function of these cellular populations. Given notable similarities in developmental gene expression between pancreatic ß-cells and serotonergic neurons, we examined the pattern of Cre-mediated recombination in the nervous system of a widely used mouse line, Pdx1-cre (formal designation, Tg(Ipf1-cre)89.1Dam), in which the expression of Cre recombinase is driven by regulatory elements upstream of the pdx1 (pancreatic-duodenal homeobox 1) gene. METHODS: Single (hemizygous) transgenic mice of the pdx1-creCre/0 genotype were bred to single (hemizygous) transgenic reporter mice (Z/EG and rosa26R lines). Recombination pattern was examined in offspring using whole-mount and sectioned histological preparations at e9.5, e10.5, e11.5, e16.5 and adult developmental stages. RESULTS: In addition to the previously reported pancreatic recombination, recombination in the developing nervous system and inner ear formation was observed. In the central nervous system, we observed a highly specific pattern of recombination in neuronal progenitors in the ventral brainstem and diencephalon. In the rostral brainstem (r1-r2), recombination occurred in newborn serotonergic neurons. In the caudal brainstem, recombination occurred in non-serotonergic cells. In the adult, this resulted in reporter expression in the vast majority of forebrain-projecting serotonergic neurons (located in the dorsal and median raphe nuclei) but in none of the spinal cord-projecting serotonergic neurons of the caudal raphe nuclei. In the adult caudal brainstem, reporter expression was widespread in the inferior olive nucleus. In the adult hypothalamus, recombination was observed in the arcuate nucleus and dorsomedial hypothalamus. Recombination was not observed in any other region of the central nervous system. Neuronal expression of endogenous pdx1 was not observed. CONCLUSIONS: The Pdx1-cre mouse line, and the regulatory elements contained in the corresponding transgene, could be a valuable tool for targeted genetic manipulation of developing forebrain-projecting serotonergic neurons and several other unique neuronal sub-populations. These results suggest that investigators employing this mouse line for studies of pancreatic function should consider the possible contributions of central nervous system effects towards resulting phenotypes.


Subject(s)
Homeodomain Proteins/genetics , Hypothalamus/cytology , Integrases/genetics , Mice, Transgenic , Neurons/physiology , Recombination, Genetic , Serotonin/metabolism , Trans-Activators/genetics , Animals , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Genes, Reporter , Genotype , Hypothalamus/physiology , Mice , Mice, Transgenic/embryology , Mice, Transgenic/physiology , Neurons/cytology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
12.
Genes Dev ; 24(20): 2330-42, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20952540

ABSTRACT

Serotonin is a bioamine regulating bone mass accrual differently depending on its site of synthesis. It decreases accrual when synthesized in the gut, and increases it when synthesized in the brain. The signal transduction events elicited by gut-derived serotonin once it binds to the Htr1b receptor present on osteoblasts have been identified and culminate in cAMP response element-binding protein (CREB) regulation of osteoblast proliferation. In contrast, we do not know how brain-derived serotonin favors bone mass accrual following its binding to the Htr2c receptor on neurons of the hypothalamic ventromedial nucleus (VMH). We show here--through gene expression analysis, serotonin treatment of wild-type and Htr2c(-/-) hypothalamic explants, and cell-specific gene deletion in the mouse--that, following its binding to the Htr2c receptor on VMH neurons, serotonin uses a calmodulin kinase (CaMK)-dependent signaling cascade involving CaMKKß and CaMKIV to decrease the sympathetic tone and increase bone mass accrual. We further show that the transcriptional mediator of these events is CREB, whose phosphorylation on Ser 133 is increased by CaMKIV following serotonin treatment of hypothalamic explants. A microarray experiment identified two genes necessary for optimum sympathetic activity whose expression is regulated by CREB. These results provide a molecular understanding of how serotonin signals in hypothalamic neurons to regulate bone mass accrual and identify CREB as a critical determinant of this function, although through different mechanisms depending on the cell type, neuron, or osteoblast in which it is expressed.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Neurons/metabolism , Osteoblasts/metabolism , Serotonin/metabolism , Animals , Bone and Bones/cytology , Bone and Bones/metabolism , Brain/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Line, Tumor , Cluster Analysis , Cyclic AMP Response Element-Binding Protein/genetics , Female , Fluorescent Antibody Technique , Gene Expression/drug effects , Gene Expression Profiling , Hypothalamus/cytology , Hypothalamus/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Phosphorylation/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Serotonin/pharmacology
13.
PLoS One ; 5(7): e11802, 2010 Jul 27.
Article in English | MEDLINE | ID: mdl-20668550

ABSTRACT

The ability to entrain circadian rhythms to food availability is important for survival. Food-entrained circadian rhythms are characterized by increased locomotor activity in anticipation of food availability (food anticipatory activity). However, the molecular components and neural circuitry underlying the regulation of food anticipatory activity remain unclear. Here we show that serotonin(2C) receptor (5-HT2CR) null mutant mice subjected to a daytime restricted feeding schedule exhibit enhanced food anticipatory activity compared to wild-type littermates, without phenotypic differences in the impact of restricted feeding on food consumption, body weight loss, or blood glucose levels. Moreover, we show that the enhanced food anticipatory activity in 5-HT2CR null mutant mice develops independent of external light cues and persists during two days of total food deprivation, indicating that food anticipatory activity in 5-HT2CR null mutant mice reflects the locomotor output of a food-entrainable oscillator. Whereas restricted feeding induces c-fos expression to a similar extent in hypothalamic nuclei of wild-type and null mutant animals, it produces enhanced expression in the nucleus accumbens and other extrahypothalamic regions of null mutant mice relative to wild-type subjects. These data suggest that 5-HT2CRs gate food anticipatory activity through mechanisms involving extrahypothalamic neural pathways.


Subject(s)
Feeding Behavior/physiology , Neural Pathways/metabolism , Receptor, Serotonin, 5-HT2C/physiology , Analysis of Variance , Animals , Blood Glucose , Body Weight/genetics , Body Weight/physiology , Eating/genetics , Eating/physiology , Food Deprivation/physiology , In Situ Hybridization , Male , Mice , Mice, Knockout , Motor Activity , Proto-Oncogene Proteins c-fos/genetics , Receptor, Serotonin, 5-HT2C/genetics
14.
Nat Med ; 16(7): 804-8, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20581837

ABSTRACT

During pregnancy, the energy requirements of the fetus impose changes in maternal metabolism. Increasing insulin resistance in the mother maintains nutrient flow to the growing fetus, whereas prolactin and placental lactogen counterbalance this resistance and prevent maternal hyperglycemia by driving expansion of the maternal population of insulin-producing beta cells. However, the exact mechanisms by which the lactogenic hormones drive beta cell expansion remain uncertain. Here we show that serotonin acts downstream of lactogen signaling to stimulate beta cell proliferation. Expression of serotonin synthetic enzyme tryptophan hydroxylase-1 (Tph1) and serotonin production rose sharply in beta cells during pregnancy or after treatment with lactogens in vitro. Inhibition of serotonin synthesis by dietary tryptophan restriction or Tph inhibition blocked beta cell expansion and induced glucose intolerance in pregnant mice without affecting insulin sensitivity. Expression of the G alpha(q)-linked serotonin receptor 5-hydroxytryptamine receptor-2b (Htr2b) in maternal islets increased during pregnancy and normalized just before parturition, whereas expression of the G alpha(i)-linked receptor Htr1d increased at the end of pregnancy and postpartum. Blocking Htr2b signaling in pregnant mice also blocked beta cell expansion and caused glucose intolerance. These studies reveal an integrated signaling pathway linking beta cell mass to anticipated insulin need during pregnancy. Modulators of this pathway, including medications and diet, may affect the risk of gestational diabetes.


Subject(s)
Insulin-Secreting Cells/metabolism , Pregnancy, Animal , Serotonin/metabolism , Animals , Female , Gene Expression Profiling , Glucose/metabolism , Insulin/metabolism , Insulin-Secreting Cells/physiology , Islets of Langerhans/physiology , Mice , Mice, Inbred C57BL , Placental Lactogen/metabolism , Pregnancy , Prolactin/metabolism , Tryptophan Hydroxylase/metabolism
15.
Am J Physiol Lung Cell Mol Physiol ; 299(2): L272-80, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20495077

ABSTRACT

Serotonin (5-HT), known as neuromodulator, regulates immune responses and inflammatory cascades. The expression and function of 5-HT receptors on alveolar macrophages (AM), which are the major fraction of pulmonary immune cells, remain elusive. Therefore, we determined the expression of 5-HT type 2 receptors and investigated the effects evoked by stimulation with 5-HT in AM compared with alveolar epithelial cells (AEC). Quantitative PCR (qPCR) analysis revealed expression of the receptors 5-HT(2A) and 5-HT(2B) in AEC and of 5-HT(2C) in AM. In AM, 5-HT (10(-5) M) induced a rise in intracellular calcium concentration ([Ca(2+)](i)) that was initiated by release of Ca(2+) from intracellular stores and depended on extracellular Ca(2+) in a sustained phase. This 5-HT-induced increase in [Ca(2+)](i) was not observed in AM treated with the 5-HT(2C)-selective inhibitor RS-102221 and in AM derived from 5-HT(2C)-deficient mice. AM stimulated with 5-HT (10(-5) M) showed increased expression of CCL2 (MCP-1) mRNA as assayed by qPCR at 4 h and augmented production of CCL2 protein as determined by dot-blot assay and ELISA at 24 h. Notably, in 5-HT(2C)-deficient AM, CCL2 production was not induced by 5-HT treatment. Moreover, transcriptional responses to 5-HT exposure assayed by microarray experiments were only observed in AM from wild-type animals and not in AM derived from 5-HT(2C)-deficient mice. Taken together, these data demonstrate the presence of functional 5-HT(2C) receptors on AM and suggest a role of 5-HT as novel modulator of AM function. These effects are exclusively driven by the 5-HT(2C) receptor, thereby providing the potential for selective intervention.


Subject(s)
Macrophages, Alveolar/metabolism , Receptor, Serotonin, 5-HT2C/physiology , Serotonin/pharmacology , Animals , Calcium/metabolism , Chemokine CCL2/biosynthesis , Mice , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism , Serotonin 5-HT2 Receptor Antagonists , Spiro Compounds/pharmacology , Sulfonamides/pharmacology
16.
J Neurodev Disord ; 1(3): 224-36, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19816534

ABSTRACT

UNLABELLED: The inhibitory GABAergic system has been implicated in multiple neuropsychiatric diseases such as schizophrenia and autism. The Dlx homeobox transcription factor family is essential for development and function of GABAergic interneurons. Mice lacking the Dlx1 gene have postnatal subtype-specific loss of interneurons and reduced IPSCs in their cortex and hippocampus. To ascertain consequences of these changes in the GABAergic system, we performed a battery of behavioral assays on the Dlx1 mutant mice, including zero maze, open field, locomotor activity, food intake, rotarod, tail suspension, fear conditioning assays (context and trace), prepulse inhibition, and working memory related tasks (spontaneous alteration task and spatial working memory task). Dlx1 mutant mice displayed elevated activity levels in open field, locomotor activity, and tail suspension tests. These mice also showed deficits in contextual and trace fear conditioning, and possibly in prepulse inhibition. Their learning deficits were not global, as the mutant mice did not differ from the wild-type controls in tests of working memory. Our findings demonstrate a critical role for the Dlx1 gene, and likely the subclasses of interneurons that are affected by the lack of this gene, in behavioral inhibition and associative fear learning. These observations support the involvement of particular components of the GABAergic system in specific behavioral phenotypes related to complex neuropsychiatric diseases. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1007/s11689-009-9025-8) contains supplementary material, which is available to authorized users.

17.
Cell ; 138(5): 976-89, 2009 Sep 04.
Article in English | MEDLINE | ID: mdl-19737523

ABSTRACT

Leptin inhibition of bone mass accrual requires the integrity of specific hypothalamic neurons but not expression of its receptor on these neurons. The same is true for its regulation of appetite and energy expenditure. This suggests that leptin acts elsewhere in the brain to achieve these three functions. We show here that brainstem-derived serotonin (BDS) favors bone mass accrual following its binding to Htr2c receptors on ventromedial hypothalamic neurons and appetite via Htr1a and 2b receptors on arcuate neurons. Leptin inhibits these functions and increases energy expenditure because it reduces serotonin synthesis and firing of serotonergic neurons. Accordingly, while abrogating BDS synthesis corrects the bone, appetite and energy expenditure phenotypes caused by leptin deficiency, inactivation of the leptin receptor in serotonergic neurons recapitulates them fully. This study modifies the map of leptin signaling in the brain and identifies a molecular basis for the common regulation of bone and energy metabolisms. For a video summary of this article, see the PaperFlick file with the Supplemental Data available online.


Subject(s)
Appetite , Bone Density , Energy Metabolism , Leptin/metabolism , Serotonin/metabolism , Brain Stem/metabolism , Hypothalamus/metabolism , Receptors, Leptin/metabolism , Signal Transduction
18.
PLoS One ; 4(8): e6797, 2009 Aug 27.
Article in English | MEDLINE | ID: mdl-19710918

ABSTRACT

BACKGROUND: Serotonin (5-HT) is a neurotransmitter with important roles in the regulation of neurobehavioral processes, particularly those regulating affect in humans. Drugs that potentiate serotonergic neurotransmission by selectively inhibiting the reuptake of serotonin (SSRIs) are widely used for the treatment of psychiatric disorders. Although the regulation of serotonin synthesis may be an factor in SSRI efficacy, the effect of chronic SSRI administration on 5-HT synthesis is not well understood. Here, we describe effects of chronic administration of the SSRI citalopram (CIT) on 5-HT synthesis and content in the mouse forebrain. METHODOLOGY/PRINCIPAL FINDINGS: Citalopram was administered continuously to adult male C57BL/6J mice via osmotic minipump for 2 days, 14 days or 28 days. Plasma citalopram levels were found to be within the clinical range. 5-HT synthesis was assessed using the decarboxylase inhibition method. Citalopram administration caused a suppression of 5-HT synthesis at all time points. CIT treatment also caused a reduction in forebrain 5-HIAA content. Following chronic CIT treatment, forebrain 5-HT stores were more sensitive to the depleting effects of acute decarboxylase inhibition. CONCLUSIONS/SIGNIFICANCE: Taken together, these results demonstrate that chronic citalopram administration causes a sustained suppression of serotonin synthesis in the mouse forebrain. Furthermore, our results indicate that chronic 5-HT reuptake inhibition renders 5-HT brain stores more sensitive to alterations in serotonin synthesis. These results suggest that the regulation of 5-HT synthesis warrants consideration in efforts to develop novel antidepressant strategies.


Subject(s)
Antidepressive Agents, Second-Generation/pharmacology , Citalopram/pharmacology , Prosencephalon/drug effects , Serotonin/biosynthesis , Animals , Antidepressive Agents, Second-Generation/blood , Citalopram/blood , Male , Mice , Mice, Inbred C57BL , Prosencephalon/metabolism
19.
J Neurosci ; 29(25): 8156-65, 2009 Jun 24.
Article in English | MEDLINE | ID: mdl-19553455

ABSTRACT

The impact of serotonergic neurotransmission on brain dopaminergic pathways has substantial relevance to many neuropsychiatric disorders. A particularly prominent role has been ascribed to the inhibitory effects of serotonin 2C receptor (5-HT(2C)R) activation on physiology and behavior mediated by the mesolimbic dopaminergic pathway, particularly in the terminal region of the nucleus accumbens. The influence of this receptor subtype on functions mediated by the nigrostriatal dopaminergic pathway is less clear. Here we report that a null mutation eliminating expression of 5-HT(2C)Rs produces marked alterations in the activity and functional output of this pathway. 5-HT(2C)R mutant mice displayed increased activity of substantia nigra pars compacta (SNc) dopaminergic neurons, elevated baseline extracellular dopamine concentrations in the dorsal striatum (DSt), alterations in grooming behavior, and enhanced sensitivity to the stereotypic behavioral effects of d-amphetamine and GBR 12909. These psychostimulant responses occurred in the absence of phenotypic differences in drug-induced extracellular dopamine concentration, suggesting a phenotypic alteration in behavioral responses to released dopamine. This was further suggested by enhanced behavioral responses of mutant mice to the D(1) receptor agonist SKF 81297. Differences in DSt D(1) or D(2) receptor expression were not found, nor were differences in medium spiny neuron firing patterns or intrinsic membrane properties following dopamine stimulation. We conclude that 5-HT(2C)Rs regulate nigrostriatal dopaminergic activity and function both at SNc dopaminergic neurons and at a locus downstream of the DSt.


Subject(s)
Behavior, Animal/physiology , Corpus Striatum/physiology , Dopamine/metabolism , Neural Pathways/physiology , Neurons/physiology , Receptor, Serotonin, 5-HT2C/physiology , Substantia Nigra/physiology , Amphetamine/administration & dosage , Amphetamine/pharmacology , Animals , Autoradiography , Behavior, Animal/drug effects , Benzazepines/administration & dosage , Benzazepines/pharmacology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dopamine Agents/administration & dosage , Dopamine Agents/pharmacology , Dopamine Agonists/administration & dosage , Dopamine Agonists/pharmacology , Dopamine Uptake Inhibitors/administration & dosage , Dopamine Uptake Inhibitors/pharmacology , Electrophysiology , Grooming/physiology , Locomotion/physiology , Mice , Mice, Inbred C57BL , Mutation , Neurons/drug effects , Neurons/metabolism , Piperazines/administration & dosage , Piperazines/pharmacology , Receptor, Serotonin, 5-HT2C/deficiency , Receptor, Serotonin, 5-HT2C/genetics , Stereotyped Behavior/physiology , Substantia Nigra/drug effects , Substantia Nigra/metabolism
20.
Proc Natl Acad Sci U S A ; 105(52): 20575-82, 2008 Dec 30.
Article in English | MEDLINE | ID: mdl-19106295

ABSTRACT

Patterns of behavior exhibited by mice in their home cages reflect the function and interaction of numerous behavioral and physiological systems. Detailed assessment of these patterns thus has the potential to provide a powerful tool for understanding basic aspects of behavioral regulation and their perturbation by disease processes. However, the capacity to identify and examine these patterns in terms of their discrete levels of organization across diverse behaviors has been difficult to achieve and automate. Here, we describe an automated approach for the quantitative characterization of fundamental behavioral elements and their patterns in the freely behaving mouse. We demonstrate the utility of this approach by identifying unique features of home cage behavioral structure and changes in distinct levels of behavioral organization in mice with single gene mutations altering energy balance. The robust, automated, reproducible quantification of mouse home cage behavioral structure detailed here should have wide applicability for the study of mammalian physiology, behavior, and disease.


Subject(s)
Behavior, Animal , Energy Metabolism , Animals , Energy Metabolism/genetics , Female , Male , Mice , Mice, Obese
SELECTION OF CITATIONS
SEARCH DETAIL
...